1
|
Huang J, Zhou X, Xu Y, Yu C, Zhang H, Qiu J, Wei J, Luo Q, Xu Z, Lin Y, Qiu P, Li C. Shen Qi Wan regulates OPN/CD44/PI3K pathway to improve airway inflammation in COPD: Network pharmacology, bioinformatics, and experimental validation. Int Immunopharmacol 2025; 144:113624. [PMID: 39577218 DOI: 10.1016/j.intimp.2024.113624] [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: 09/10/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is one of the most common respiratory diseases with undefined pathogenesis and unsatisfactory therapeutic options. Shenqi Wan (SQW), a traditional Chinese medicinal compound, has demonstrated certain preventive and therapeutic effects on COPD. However, the underlying molecular mechanisms remain incompletely understood. In this study, we used weighted gene co-expression network analysis (WGCNA) and machine learning to identify biomarkers for COPD, combined with network pharmacology and experimental validation to evaluate how SQW reduces airway inflammation in COPD. METHODS Targets of SQW in treating COPD and its network regulation mechanism were predicted via network pharmacology. Meanwhile, potential biomarkers were predicted using WGCNA and machine learning algorithms and validated in COPD patients. The relationship between the core pathway and key target was analyzed by ingenuity pathway analysis (IPA) to reveal the regulatory mechanism of SQW. We evaluated the efficacy of SQW treatment in LPS/MS-induced COPD mice by evaluating lung function, histopathological parameters, and levels of inflammatory markers and oxidative stress. The distribution and expression of OPN/CD44/PI3K loop-related proteins were examined through immunofluorescence staining and Western Blotting. In vitro, we added LPS to BEAS-2B cells to mimic the inflammatory microenvironment and transfected the cells with OPN overexpression plasmid to observe the improvement induced by SQW. RESULTS GO and KEGG analyses demonstrated that SQW inhibited inflammation and oxidative stress via the PI3K/Akt pathway, thereby improving COPD. Machine learning algorithms identified OPN as a potential biomarker, with elevated expression observed in the lung tissue of COPD patients. IPA indicated that OPN may modulate the CD44-mediated activation of the PI3K/AKT pathway, forming a positive feedback regulatory mechanism. SQW ameliorated lung function and pathological injury in mice; further, it reduced inflammation, oxidative stress, and OPN/CD44/PI3K positive feedback loop-related protein expression in both mice and cells. After OPN overexpression, the levels of inflammatory factors and ROS were significantly increased, and the OPN/CD44/PI3K signal was further activated, weakening the ameliorative effect of the SQW drug-containing serum. CONCLUSION Overall, SQW contributed to ameliorating COPD by reducing airway inflammation and oxidative stress through inhibiting the OPN/CD44/PI3K positive feedback loop.
Collapse
Affiliation(s)
- Junhao Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaojie Zhou
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yueling Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chenshi Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huanhuan Zhang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xian 710032, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou 311121, China
| | - Jiale Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhiwei Xu
- Jinhua Academy, Zhejiang Chinese Medical University, Jinhua 321000, China
| | - Yiyou Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ping Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
2
|
Shen D, Seco BMS, Teixeira Alves LG, Yao L, Bräutigam M, Opitz B, Witzenrath M, Fries BC, Seeberger PH. Semisynthetic Glycoconjugate Vaccine Lead against Klebsiella pneumoniae Serotype O2afg Induces Functional Antibodies and Reduces the Burden of Acute Pneumonia. J Am Chem Soc 2024; 146:35356-35366. [PMID: 39666976 DOI: 10.1021/jacs.4c13972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CR-Kp) bacteria are a serious global health concern due to their drug-resistance to nearly all available antibiotics, fast spread, and high mortality rate. O2afg is a major CR-Kp serotype in the sequence type 258 group (KPST258) that is weakly immunogenic in humans. Here, we describe the creation and evaluation of semisynthetic O2afg glycoconjugate vaccine leads containing one and two repeating units of the polysaccharide epitope that covers the surface of the bacteria conjugated to the carrier protein CRM197. The semisynthetic glycoconjugate containing two repeating units induced functional IgG antibodies in rabbits with opsonophagocytic killing activity and enhanced complement activation and complement-mediated killing of CR-Kp. Passive immunization reduced the burden of acute pneumonia in mice and may represent an alternative to antimicrobial therapy. The semisynthetic glycoconjugate vaccine lead against CR-Kp expressing O2afg antigen is awaiting preclinical development.
Collapse
Affiliation(s)
- Dacheng Shen
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bruna M S Seco
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Luiz Gustavo Teixeira Alves
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
| | - Ling Yao
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Maria Bräutigam
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bastian Opitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
- German Center for Lung Research (DZL), 12203 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
- German Center for Lung Research (DZL), 12203 Berlin, Germany
| | - Bettina C Fries
- Department of Medicine, Infectious Disease Division, Stony Brook University; Stony Brook, New York 11794, United States
- Veteran's Administration Medical Center, Northport, New York 11768, United States
| | - Peter H Seeberger
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
3
|
Hamamoto K, Liang X, Ito A, Lanza M, Bui V, Zhang J, Opozda DM, Hattori T, Chen L, Haddock D, Imamura F, Wang HG, Takahashi Y. Unveiling the physiological impact of ESCRT-dependent autophagosome closure by targeting the VPS37A ubiquitin E2 variant-like domain. Cell Rep 2024; 43:115016. [PMID: 39607828 DOI: 10.1016/j.celrep.2024.115016] [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: 01/08/2024] [Revised: 09/05/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Macroautophagy (autophagy) involves the formation of phagophores that mature into autophagosomes. The impact of inhibiting autophagosome closure remains unclear. Here, we report the generation and analysis of mice with impaired autophagosome closure by targeting the ubiquitin E2 variant-like (UEVL) β strands of the endosomal sorting complex required for transport (ESCRT) I subunit VPS37A. The VPS37A UEVL mutation (Δ43-139) impairs bulk autophagic flux without disrupting ESCRT-I complex assembly and endosomal function. Homozygous mutant mice exhibit signs of autophagy impairment, including p62/SQSTM1 and ubiquitinated protein accumulation, neuronal dysfunction, growth retardation, antioxidant gene upregulation, and tissue abnormalities. However, about half of the mutant neonates survive to adulthood without severe liver injury. LC3 proximity proteomics reveals that the VPS37A UEVL mutation leads to active TANK-binding kinase 1 (TBK1) accumulation on phagophores, resulting in increased p62 phosphorylation and inclusion formation. These findings reveal a previously unappreciated role of LC3-conjugated phagophores in facilitating protein aggregation and sequestration, potentially alleviating proteotoxicity.
Collapse
Affiliation(s)
- Kouta Hamamoto
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Xinwen Liang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ayako Ito
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Matthew Lanza
- Department of Comparative Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Van Bui
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jiawen Zhang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - David M Opozda
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Tatsuya Hattori
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Longgui Chen
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - David Haddock
- Department of Pathology and Biochemistry, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Fumiaki Imamura
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Hong-Gang Wang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Yoshinori Takahashi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
4
|
Zaparina OG, Kapushchak YK, Lishai EA, Hong SJ, Sripa B, Pakharukova MY. Species-specific renal and liver responses during infection with food-borne trematodes Opisthorchis felineus, Opisthorchis viverrini, or Clonorchis sinensis. PLoS One 2024; 19:e0311481. [PMID: 39637122 PMCID: PMC11620611 DOI: 10.1371/journal.pone.0311481] [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: 06/13/2024] [Accepted: 09/03/2024] [Indexed: 12/07/2024] Open
Abstract
Three food-borne trematodes-Opisthorchis felineus, Opisthorchis viverrini, and Clonorchis sinensis-are closely related epidemiologically important species. Despite the similarity of their life cycles, these liver flukes also have marked differences in the geographical range, helminth biology, and hepatobiliary disorders. O. viverrini and C. sinensis are classified as Group 1 biological carcinogens while O. felineus is not. Direct comparisons of systemic response to the liver fluke infections are unexplored aspects. This study was carried out to identify species-specific liver and kidney responses in the hamster models after the infection with one of the three liver flukes. Liver periductal-fibrosis development was similar between hamsters infected with O. felineus or C. sinensis, whereas biliary intraepithelial neoplasia development was noticed predominantly in O. viverrini-infected ones. Species-specific renal damage was detected, including progression of interstitial fibrosis and IgA deposition in glomeruli of O. felineus-infected hamsters and C. sinensis-infected ones. A strong correlation (R = 0.63; P = 0.0001) was found between periductal fibrosis in the liver and kidney interstitial fibrosis. Future comparative studies are needed to elucidate the development of serious complications during the long term of the infection, as well as under the influence of additional factors, including concomitant infections and the use of dimethylnitrosamine to clarify the mechanisms underlying the liver fluke-associated carcinogenesis. Thus, our findings may stimulate new comparative studies on the pathogenicity.
Collapse
Affiliation(s)
- Oxana G. Zaparina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
| | - Yaroslav K. Kapushchak
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
| | - Ekaterina A. Lishai
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Sung-Jong Hong
- Center for Infectious Diseases and Vectors, Incheon National University, Incheon, Korea
| | - Banchob Sripa
- Faculty of Medicine, Department of Pathology, WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - Maria Y. Pakharukova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (ICG SB RAS), Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
5
|
Gonzalez-Orozco M, Tseng HC, Hage A, Xia H, Behera P, Afreen K, Peñaflor-Tellez Y, Giraldo MI, Huante M, Puebla-Clark L, van Tol S, Odle A, Crown M, Teruel N, Shelite TR, Moreno-Contreras J, Terasaki K, Makino S, Menachery V, Endsley M, Endsley JJ, Najmanovich RJ, Bashton M, Stephens R, Shi PY, Xie X, Freiberg AN, Rajsbaum R. TRIM7 ubiquitinates SARS-CoV-2 membrane protein to limit apoptosis and viral replication. Nat Commun 2024; 15:10438. [PMID: 39616206 PMCID: PMC11608229 DOI: 10.1038/s41467-024-54762-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024] Open
Abstract
SARS-CoV-2 is a highly transmissible virus that causes COVID-19 disease. Mechanisms of viral pathogenesis include excessive inflammation and viral-induced cell death, resulting in tissue damage. Here we show that the host E3-ubiquitin ligase TRIM7 acts as an inhibitor of apoptosis and SARS-CoV-2 replication via ubiquitination of the viral membrane (M) protein. Trim7-/- mice exhibit increased pathology and virus titers associated with epithelial apoptosis and dysregulated immune responses. Mechanistically, TRIM7 ubiquitinates M on K14, which protects cells from cell death. Longitudinal SARS-CoV-2 sequence analysis from infected patients reveal that mutations on M-K14 appeared in circulating variants during the pandemic. The relevance of these mutations was tested in a mouse model. A recombinant M-K14/K15R virus shows reduced viral replication, consistent with the role of K15 in virus assembly, and increased levels of apoptosis associated with the loss of ubiquitination on K14. TRIM7 antiviral activity requires caspase-6 inhibition, linking apoptosis with viral replication and pathology.
Collapse
Affiliation(s)
- Maria Gonzalez-Orozco
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hsiang-Chi Tseng
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Adam Hage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hongjie Xia
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Padmanava Behera
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Kazi Afreen
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Yoatzin Peñaflor-Tellez
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Maria I Giraldo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Matthew Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lucinda Puebla-Clark
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Sarah van Tol
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Abby Odle
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Matthew Crown
- Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Natalia Teruel
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Thomas R Shelite
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Joaquin Moreno-Contreras
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Kaori Terasaki
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Shinji Makino
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Vineet Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mark Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rafael J Najmanovich
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Matthew Bashton
- Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Ricardo Rajsbaum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
6
|
Ramadan RM, Bakr AF, Fouad E, Mohammed FF, Abdel-Wahab AM, Abdel-Maogood SZ, El-Bahy MM, Salem MA. Novel insights into antioxidant status, gene expression, and immunohistochemistry in an animal model infected with camel-derived Trypanosoma evansi and Theileria annulata. Parasit Vectors 2024; 17:474. [PMID: 39558410 PMCID: PMC11575088 DOI: 10.1186/s13071-024-06564-3] [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: 07/07/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Hemoprotozoan diseases, especially trypanosomosis and theileriosis, adversely affect the productivity, growth, and performance of camels. Regular sampling and investigation of camels are challenging due to several factors. Consequently, there is a lack of knowledge on camel parasite genotyping, cytokine production, and oxidative stress parameters during infection. METHODS The present study investigated two critical blood protozoa infecting camels in Egypt, Trypanosoma evansi and Theileria annulata, using molecular methods, specifically 18S rRNA gene analysis. Following molecular confirmation, experimental infections were induced in Swiss albino mice to assess the expression of immune response genes and oxidative stress parameters. The study further explored the correlation between histopathological alterations and inflammatory reactions in the kidney, spleen, and liver of infected mice, alongside the immunohistochemical expression of caspase-3, proliferating cell nuclear antigen (PCNA), and tumor necrosis factor (TNF). RESULTS Trypanosoma evansi and T. annulata isolated from naturally infected camels were molecularly identified and deposited in GenBank under accession numbers OR116429 and OR103130, respectively. Infection with T. evansi and T. annulata caused significant adverse effects on the immune condition of infected mice, increasing the pathogenicity of the infection. This was evidenced by a significant increase in oxidative stress parameter levels in both naturally infected camels and experimentally infected mice compared to healthy controls. Furthermore, the expression of immune response genes was significantly elevated in infected mice. Immunohistochemistry analysis showed a pronounced upregulation of caspase-3, PCNA, and TNF in the infected groups relative to the control group. These findings are the first to be reported in Egypt. CONCLUSIONS This study successfully identified and genotyped two economically important blood protozoa, T. evansi and T. annulata, from camels in Egypt. Additionally, the experimental animal model provided valuable insights into the immune response, oxidative stress, and histopathological changes induced by these parasites, demonstrating comparable results to naturally infected camels. These findings highlight the potential of this model to study parasite-host interactions and immune responses, contributing to a better understanding of the pathogenic mechanisms of T. evansi and T. annulata infections. This model may be useful for future studies focused on disease control and therapeutic interventions.
Collapse
Affiliation(s)
- Reem M Ramadan
- Department of Parasitology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt.
| | - Alaa F Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt
| | - Esraa Fouad
- The Central Laboratory for Evaluation of Veterinary Biologics (CLEVB), Agriculture Research Centre (ARC), Cairo, Egypt
| | - Faten F Mohammed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt
- Department of Pathology, College of Veterinary Medicine, King Faisal University, 31982, Al-Ahsa, Saudi Arabia
| | - Azza M Abdel-Wahab
- Department of Parasitology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt
| | - Sahar Z Abdel-Maogood
- Department of Parasitology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt
| | - Mohamed M El-Bahy
- Department of Parasitology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt
| | - Mai A Salem
- Department of Parasitology, Faculty of Veterinary Medicine, Cairo University, Giza, 1221, Egypt
| |
Collapse
|
7
|
Wu J, Zeng W, Xie H, Cao M, Yang J, Xie Y, Luo Z, Zhang Z, Xu H, Huang W, Zhou T, Tan J, Wu X, Yang Z, Zhu S, Mao R, He Z, Lan P. Microbiota-induced alteration of kynurenine metabolism in macrophages drives formation of creeping fat in Crohn's disease. Cell Host Microbe 2024; 32:1927-1943.e9. [PMID: 39541945 DOI: 10.1016/j.chom.2024.10.008] [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: 05/26/2024] [Revised: 08/28/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024]
Abstract
Hyperplasia of mesenteric tissues in Crohn's disease, called creeping fat (CrF), is associated with surgical recurrence. Although microbiota translocation and colonization have been found in CrF, convincing mouse phenotypes and the underlying mechanisms of CrF formation remain unclear. Utilizing single-nucleus RNA (snRNA) sequencing of CrF and different mouse models, we demonstrate that the commensal Achromobacter pulmonis induces mesenteric adipogenesis through macrophage alteration. Targeted metabolome analysis reveals that L-kynurenine is the most enriched metabolite in CrF. Upregulation of indoleamine 2,3-dioxygenase 1 (IDO1) enhances kynurenine metabolism and drives mesenteric adipogenesis. Leveraging single-cell RNA (scRNA) sequencing of mouse mesenteric tissues and macrophage-specific IDO1 knockout mice, we verify the role of macrophage-sourced L-kynurenine in mesenteric adipogenesis. Mechanistically, L-kynurenine-induced adipogenesis is mediated by the aryl hydrocarbon receptors in adipocytes. Administration of an IDO1 inhibitor or bacteria engineered to degrade L-kynurenine alleviates mesenteric adipogenesis in mice. Collectively, our study demonstrates that microbiota-induced modulation of macrophage metabolism potentiates CrF formation.
Collapse
Affiliation(s)
- Jinjie Wu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wanyi Zeng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Hongyu Xie
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Anesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mujia Cao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Jingyi Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Yanchun Xie
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zhanhao Luo
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Zongjin Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haoyang Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Weidong Huang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tingyue Zhou
- Key Laboratory of immune response and immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jinyu Tan
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaomin Wu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zihuan Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Shu Zhu
- Key Laboratory of immune response and immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Zhen He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China.
| | - Ping Lan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; State Key Laboratory of Oncology in South China, Guangzhou, China.
| |
Collapse
|
8
|
Bu HF, Subramanian S, Chou PM, Liu F, Sun L, Geng H, Wang X, Liao J, Du C, Hu J, Tan SC, Nathan N, Yang GY, Tan XD. A novel mouse model of hepatocyte-specific apoptosis-induced myeloid cell-dominant sterile liver injury and repair response. Am J Physiol Gastrointest Liver Physiol 2024; 327:G499-G512. [PMID: 39104322 PMCID: PMC11482258 DOI: 10.1152/ajpgi.00005.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/07/2024]
Abstract
Apoptosis, inflammation, and wound healing are critical pathophysiological events associated with various liver diseases. Currently, there is a lack of in vivo approaches to study hepatocyte apoptosis-induced liver injury and repair. To address this critical knowledge gap, we developed a unique genetically modified mouse model, namely, 3-Transgene (Tg) with inducible Hepatocyte-Specific Apoptosis Phenotype (3xTg-iHAP) in this study. The 3xTg-iHAP mice possess three transgenes including Alb-Cre, Rosa26-rtTA, and tetO-Fasl on a B6 background. These mice are phenotypically normal, viable, and fertile. After subcutaneous administration of a single dose of doxycycline (5 mg/kg, Dox) to 3xTg-iHAP mice, we observed a complete histological spectrum of sterile liver wound-healing responses: asymptomatic hepatocyte apoptosis at 8 h, necrotic liver injury and sterile inflammation at 48 h, followed by hepatocyte mitosis and regeneration within 7 days. During the injury phase, the mice exhibited an increase in the biomarkers of alanine aminotransferase (ALT), chemokine (C-X-C motif) ligand 1 (CXCL1), and IL-6 in peripheral blood, as well as α-smooth muscle actin (α-SMA) protein in liver tissues. Conversely, the mice displayed a decrease in these markers in the recovery phase. Remarkably, this model shows that the sterile liver injury following elevated hepatocyte apoptosis is associated with an increase in myeloid cells in the liver. Within 7 days post-Dox administration, the liver of Dox-treated 3xTg-iHAP mice displays a normal histological structure, indicating the completion of wound healing. Together, we established a novel mouse model of injury and regeneration induced by hepatocyte apoptosis. This tool provides a robust in vivo platform for studying the pathophysiology of sterile liver inflammation, regeneration, and new therapeutic interventions for liver diseases.NEW & NOTEWORTHY Bu et al. present a triple-transgenic mouse model, namely, 3xTg-iHAP mice that are engineered to explore hepatocyte apoptosis-triggered sterile liver injury and regeneration. This model demonstrates a full spectrum of liver wound-healing responses from asymptomatic apoptosis to injury, myeloid cell-dominant sterile inflammation, and repair after induction of hepatocyte-specific apoptosis. The robust nature of this model makes it an invaluable in vivo tool for studying sterile liver inflammation, regeneration, and new therapeutic strategies.
Collapse
Affiliation(s)
- Heng-Fu Bu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Saravanan Subramanian
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Pauline M Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Fangyi Liu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Leyu Sun
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Hua Geng
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Xiao Wang
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| | - Jie Liao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Chao Du
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Joyce Hu
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Stephanie C Tan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Nirmal Nathan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Xiao-Di Tan
- Department of Pediatrics, Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States
| |
Collapse
|
9
|
Maldonado-Rengel R, Sócola-Barsallo Z, Vásquez B. Alterations of Liver Morphology in Senescent Rats. Int J Mol Sci 2024; 25:9846. [PMID: 39337332 PMCID: PMC11431848 DOI: 10.3390/ijms25189846] [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: 08/14/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Age-related liver changes can have important implications for health and metabolic function. This study aimed to describe the morphoquantitative alterations of the liver in senescent rats compared to adult rats. Twelve male rats were used, divided into 6-month-old adults (group A) and 36-month-old senescent rats (group S). Morphometric and histopathological studies, quantification of collagen types I and III, and stereological analyses were performed to determine the volume density of mononucleated (VvhepM) and binucleated (VvhepB) hepatocyte nuclei, surface area density (SvhepM), and number density (NvhepM) of mononucleated hepatocyte nuclei. The findings reveal an alteration of the normal liver tissue architecture in senescent rats and the presence of inflammatory lesions and fibrosis. In addition, there was a decrease in body and liver mass and volume. Group S showed a significant reduction in VvhepM and NvhepM; however, SvhepM was significantly higher. No significant differences were noted in the percentage of binucleated hepatocytes between the two groups. This study reveals substantial morphological changes in the aging liver, with possible functional implications. More research is needed on the underlying mechanisms and their consequences at older ages.
Collapse
Affiliation(s)
- Ruth Maldonado-Rengel
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Department of Health Sciences, Universidad Técnica Particular de Loja, San Cayetano Alto, Calle París, Loja 110107, Ecuador
| | - Zaida Sócola-Barsallo
- Department of Health Sciences, Universidad Técnica Particular de Loja, San Cayetano Alto, Calle París, Loja 110107, Ecuador
| | - Bélgica Vásquez
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
- Centre of Excellence in Morphological and Surgical Studies, Universidad de La Frontera, Avenida Francisco Salazar 01145, Temuco 4811230, Chile
| |
Collapse
|
10
|
Da Silva Filho J, Herder V, Gibbins MP, Dos Reis MF, Melo GC, Haley MJ, Judice CC, Val FFA, Borba M, Tavella TA, de Sousa Sampaio V, Attipa C, McMonagle F, Wright D, de Lacerda MVG, Costa FTM, Couper KN, Marcelo Monteiro W, de Lima Ferreira LC, Moxon CA, Palmarini M, Marti M. A spatially resolved single-cell lung atlas integrated with clinical and blood signatures distinguishes COVID-19 disease trajectories. Sci Transl Med 2024; 16:eadk9149. [PMID: 39259811 DOI: 10.1126/scitranslmed.adk9149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/15/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
COVID-19 is characterized by a broad range of symptoms and disease trajectories. Understanding the correlation between clinical biomarkers and lung pathology during acute COVID-19 is necessary to understand its diverse pathogenesis and inform more effective treatments. Here, we present an integrated analysis of longitudinal clinical parameters, peripheral blood markers, and lung pathology in 142 Brazilian patients hospitalized with COVID-19. We identified core clinical and peripheral blood signatures differentiating disease progression between patients who recovered from severe disease compared with those who succumbed to the disease. Signatures were heterogeneous among fatal cases yet clustered into two patient groups: "early death" (<15 days until death) and "late death" (>15 days). Progression to early death was characterized systemically and in lung histopathological samples by rapid endothelial and myeloid activation and the presence of thrombi associated with SARS-CoV-2+ macrophages. In contrast, progression to late death was associated with fibrosis, apoptosis, and SARS-CoV-2+ epithelial cells in postmortem lung tissue. In late death cases, cytotoxicity, interferon, and T helper 17 (TH17) signatures were only detectable in the peripheral blood after 2 weeks of hospitalization. Progression to recovery was associated with higher lymphocyte counts, TH2 responses, and anti-inflammatory-mediated responses. By integrating antemortem longitudinal blood signatures and spatial single-cell lung signatures from postmortem lung samples, we defined clinical parameters that could be used to help predict COVID-19 outcomes.
Collapse
Affiliation(s)
- João Da Silva Filho
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Monique Freire Dos Reis
- Department of Education and Research, Oncology Control Centre of Amazonas State (FCECON), Manaus, Brazil
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Federal University of Amazonas, Manaus, Brazil
- Amazonas Oncology Control Center Foundation, Manaus, Brazil
| | | | - Michael J Haley
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Carla Cristina Judice
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Fernando Fonseca Almeida Val
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Mayla Borba
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Delphina Rinaldi Abdel Aziz Emergency Hospital (HPSDRA), Manaus, Brazil
| | - Tatyana Almeida Tavella
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- INSERM U1016, CNRS UMR8104, University of Paris Cité, Institut Cochin, Paris, France
| | | | - Charalampos Attipa
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Fiona McMonagle
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Glasgow Imaging Facility/School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Derek Wright
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marcus Vinicius Guimaraes de Lacerda
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Kevin N Couper
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Wuelton Marcelo Monteiro
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Christopher Alan Moxon
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- (C.A.M.)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- (M.P.)
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
- (M.M.)
| |
Collapse
|
11
|
Kim KS, Lee C, Kim HS, Gu SJ, Yoon HJ, Won SB, Lee H, Lee YS, Kim SS, Kane LP, Park EJ. TIM-3 on myeloid cells promotes pulmonary inflammation through increased production of galectin-3. Commun Biol 2024; 7:1090. [PMID: 39237613 PMCID: PMC11377825 DOI: 10.1038/s42003-024-06762-w] [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: 11/10/2023] [Accepted: 08/22/2024] [Indexed: 09/07/2024] Open
Abstract
T cell immunoglobulin and mucin-containing molecule 3 (TIM-3) exhibits unique, cell type- and context-dependent characteristics and functions. Here, we report that TIM-3 on myeloid cells plays essential roles in modulating lung inflammation. We found that myeloid cell-specific TIM-3 knock-in (FSF-TIM3/LysM-Cre+) mice have lower body weight and shorter lifespan than WT mice. Intriguingly, the lungs of FSF-TIM3/LysM-Cre+ mice display excessive inflammation and features of disease-associated pathology. We further revealed that galectin-3 levels are notably elevated in TIM-3-overexpressing lung-derived myeloid cells. Furthermore, both TIM-3 blockade and GB1107, a galectin-3 inhibitor, ameliorated lung inflammation in FSF-TIM3/LysM-Cre+/- mice. Using an LPS-induced lung inflammation model with myeloid cell-specific TIM-3 knock-out mice, we demonstrated the association of TIM-3 with both lung inflammation and galectin-3. Collectively, our findings suggest that myeloid TIM-3 is an important regulator in the lungs and that modulation of TIM-3 and galectin-3 could offer therapeutic benefits for inflammation-associated lung diseases.
Collapse
Affiliation(s)
- Ki Sun Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Chanju Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
- Immuno-oncology Branch, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Hyung-Seok Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Su Jeong Gu
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Hee Jung Yoon
- Immuno-oncology Branch, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Su Bin Won
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
- Immuno-oncology Branch, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Ho Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Sang Soo Kim
- Radiological Science Branch, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Eun Jung Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea.
- Immuno-oncology Branch, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea.
| |
Collapse
|
12
|
Yu F, Ma J, Xu K, Tang Y, Wu B, Gu W, Shi Y. Effects of liver X receptor in O3-induced airway inflammation and remodeling in mice. J Thorac Dis 2024; 16:5005-5017. [PMID: 39268130 PMCID: PMC11388256 DOI: 10.21037/jtd-23-1820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/21/2024] [Indexed: 09/15/2024]
Abstract
Background The current clinical treatment of chronic obstructive pulmonary disease (COPD) mainly uses drugs to improve symptoms, but these drugs cannot reverse the progression of the disease and the pathological changes in lung tissue. This study aimed to investigate the effects and mechanisms of Liver X receptors (LXRs) in ozone (O3)-induced airway inflammation and remodeling in mice. Methods Wild mice and LXR deficient mice were exposed to O3 twice a week for 6 weeks. Some wild mice were intraperitoneally injected with T0901317 (a LXR agonist) before O3 exposure. Wild mice were exposed to ambient air and intraperitoneally injected with normal saline (NS) as control group. The lung tissues and bronchoalveolar lavage fluid (BALF) were collected to evaluate airway inflammation, airway remodeling and lipid disorder. Results After O3 exposure, LXR deficient mice showed severe airway inflammation and airway remodeling compared with the wild mice. There were a lot of foamy macrophages appeared in BALF of LXR deficient mice. The inflammatory proteins such as myeloid differentiation primary response protein 88 (MyD88) and interleukin-1 receptor-associated kinase (IRAK) in the lung tissues of LXR deficient mice were significantly increased compared with the wild mice. In wild mice exposed to O3, T0901317 treatment can alleviate airway inflammation, airway remodeling and foamy macrophages in BALF. And MyD88 and IRAK expression in lung tissue were also attenuated by T0901317 treatment. Conclusions LXRs play protective roles in O3-induced lipid accumulation, airway inflammation and airway remodeling.
Collapse
Affiliation(s)
- Fenfang Yu
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiyong Ma
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Xu
- Department of Respiratory Medicine, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing, China
| | - Yu Tang
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bining Wu
- Department of Respiratory Medicine, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing, China
| | - Wei Gu
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ying Shi
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Yaseen F, Taj M, Ravindran R, Zaffar F, Luciw PA, Ikram A, Zafar SI, Gill T, Hogarth M, Khan IH. An exploratory deep learning approach to investigate tuberculosis pathogenesis in nonhuman primate model: Combining automated radiological analysis with clinical and biomarkers data. J Med Primatol 2024; 53:e12722. [PMID: 38949157 DOI: 10.1111/jmp.12722] [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: 05/03/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Tuberculosis (TB) kills approximately 1.6 million people yearly despite the fact anti-TB drugs are generally curative. Therefore, TB-case detection and monitoring of therapy, need a comprehensive approach. Automated radiological analysis, combined with clinical, microbiological, and immunological data, by machine learning (ML), can help achieve it. METHODS Six rhesus macaques were experimentally inoculated with pathogenic Mycobacterium tuberculosis in the lung. Data, including Computed Tomography (CT), were collected at 0, 2, 4, 8, 12, 16, and 20 weeks. RESULTS Our ML-based CT analysis (TB-Net) efficiently and accurately analyzed disease progression, performing better than standard deep learning model (LLM OpenAI's CLIP Vi4). TB-Net based results were more consistent than, and confirmed independently by, blinded manual disease scoring by two radiologists and exhibited strong correlations with blood biomarkers, TB-lesion volumes, and disease-signs during disease pathogenesis. CONCLUSION The proposed approach is valuable in early disease detection, monitoring efficacy of therapy, and clinical decision making.
Collapse
Affiliation(s)
- Faisal Yaseen
- Department of Biomedical and Health Informatics, University of Washington, Seattle, Washington, USA
| | - Murtaza Taj
- Department of Computer Science, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Resmi Ravindran
- Department of Pathology and Laboratory Medicine, University of California, Sacramento, California, USA
| | - Fareed Zaffar
- Department of Computer Science, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Paul A Luciw
- Department of Pathology and Laboratory Medicine, University of California, Sacramento, California, USA
| | - Aamer Ikram
- National Institutes of Health, Islamabad, Pakistan
| | - Saerah Iffat Zafar
- Armed Forces Institute of Radiology and Imaging (AFIRI), Rawalpindi, Pakistan
| | - Tariq Gill
- Albany Medical Center, Albany, New York, USA
| | - Michael Hogarth
- Department of Medicine, University of California, San Diego, California, USA
| | - Imran H Khan
- Department of Pathology and Laboratory Medicine, University of California, Sacramento, California, USA
| |
Collapse
|
14
|
Kozień Ł, Policht A, Heczko P, Arent Z, Bracha U, Pardyak L, Pietsch-Fulbiszewska A, Gallienne E, Piwowar P, Okoń K, Tomusiak-Plebanek A, Strus M. PDIA iminosugar influence on subcutaneous Staphylococcus aureus and Pseudomonas aeruginosa infections in mice. Front Cell Infect Microbiol 2024; 14:1395577. [PMID: 39145303 PMCID: PMC11322076 DOI: 10.3389/fcimb.2024.1395577] [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: 03/04/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction Biofilm-associated infections persist as a therapeutic challenge in contemporary medicine. The efficacy of antibiotic therapies is ineffective in numerous instances, necessitating a heightened focus on exploring novel anti-biofilm medical strategies. Among these, iminosugars emerge as a distinctive class of compounds displaying promising biofilm inhibition properties. Methods This study employs an in vivo wound infection mouse model to evaluate the effectiveness of PDIA in treating biofilm-associated skin wound infections caused by Staphylococcus aureus and Pseudomonas aeruginosa. Dermic wounds in mice were infected with biofilm-forming strains, specifically S. aureus 48 and P. aeruginosa 5, which were isolated from patients with diabetic foot, and are well-known for their strong biofilm formation. The subsequent analysis included clinical, microbiological, and histopathological parameters. Furthermore, an exploration into the susceptibility of the infectious strains to hydrogen peroxide was conducted, acknowledging its potential presence during induced inflammation in mouse dermal wounds within an in vivo model. Results The findings revealed the efficacy of PDIA iminosugar against the S. aureus strain, evidenced by a reduction in bacterial numbers within the wound and the inflammatory focus. Discussion This study suggests that PDIA iminosugar emerges as an active and potentially effective antibiofilm agent, positioning it as a viable treatment option for staphylococcal infections.
Collapse
Affiliation(s)
- Łucja Kozień
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Aleksandra Policht
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Piotr Heczko
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Zbigniew Arent
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Urszula Bracha
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Laura Pardyak
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Agnieszka Pietsch-Fulbiszewska
- Center of Experimental and Innovative Medicine, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Krakow, Poland
| | - Estelle Gallienne
- Institut de Chimie Organique et Analytique (ICOA), UMR 7311, Université d'Orléans & CNRS, Orléans, France
| | - Piotr Piwowar
- Faculty of Electrical Engineering, Automatics, Computer Science and Biomedical Engineering, AGH University of Science and Technology, Kraków, Poland
| | - Krzysztof Okoń
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Tomusiak-Plebanek
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Magdalena Strus
- Department of Bacteriology, Ecology of Microbes and Parasitology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
15
|
Li Y, Wang W, Liu Y, Li S, Wang J, Hou L. Diminished Immune Response and Elevated Abundance in Gut Microbe Dubosiella in Mouse Models of Chronic Colitis with GBP5 Deficiency. Biomolecules 2024; 14:873. [PMID: 39062588 PMCID: PMC11274912 DOI: 10.3390/biom14070873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Guanylate binding protein 5 (GBP5) is an emerging immune component that has been increasingly recognized for its involvement in autoimmune diseases, particularly inflammatory bowel disease (IBD). IBD is a complex disease involving inflammation of the gastrointestinal tract. Here, we explored the functional significance of GBP5 using Gbp5 knockout mice and wildtype mice exposed to dextran sulfate sodium (DSS) to generate chronic colitis model. We found that Gbp5 deficiency protected mice from DSS-induced chronic colitis. Transcriptome analysis of colon tissues showed reduced immune responses in Gbp5 knockout mice compared to those in corresponding wildtype mice. We further observed that after repeated DSS exposure, the gut microbiota was altered, both in wildtype mice and Gbp5 knockout mice; however, the gut microbiome health index was higher in the Gbp5 knockout mice. Notably, a probiotic murine commensal bacterium, Dubosiella, was predominantly enriched in these knockout mice. Our findings suggest that GBP5 plays an important role in promoting inflammation and dysbiosis in the intestine, the prevention of which might therefore be worth exploring in regards to IBD treatment.
Collapse
Affiliation(s)
- Yichen Li
- Medical College, Jiaying University, Meizhou 514031, China
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China;
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, Department of General Surgery, The Six Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Wenxia Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China;
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Biomedical Innovation Center, Department of General Surgery, The Six Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yuxuan Liu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Y.L.); (S.L.); (J.W.)
| | - Senru Li
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Y.L.); (S.L.); (J.W.)
| | - Jingyu Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Y.L.); (S.L.); (J.W.)
| | - Linlin Hou
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (Y.L.); (S.L.); (J.W.)
| |
Collapse
|
16
|
Tan S, Li Q, Guo C, Chen S, Kamal-Eldin A, Chen G. Reveal the mechanism of hepatic oxidative stress in mice induced by photo-oxidation milk using multi-omics analysis techniques. J Adv Res 2024:S2090-1232(24)00271-6. [PMID: 38986809 DOI: 10.1016/j.jare.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
INTRODUCTION Photo-oxidation is recognized as a contributor to the deterioration of milk quality, posing potential safety hazards to human health. However, there has been limited investigation into the impact of consuming photo-oxidized milk on health. OBJECTIVES This study employs multi-omics analysis techniques to elucidate the mechanisms by which photo-oxidized milk induces oxidative stress in the liver. METHODS Mouse model was used to determine the effect of the gavage administration of milk with varying degrees of photo-oxidation on the mouse liver. The damage degree was established by measuring serum markers indicative of oxidative stress, and with a subsequent histopathological examination of liver tissues. In addition, comprehensive metabolome, lipidome, and transcriptome analyses were conducted to elucidate the underlying molecular mechanisms of hepatic damage caused by photo-oxidized milk. RESULTS A significant elevation in the oxidative stress levels and the presence of hepatocellular swelling and inflammation subsequent to the gavage administration of photo-oxidized milk to mice. Significant alterations in the levels of metabolites such as lumichrome, all-trans-retinal, L-valine, phosphatidylglycerol, and phosphatidylcholine within the hepatic tissue of mice. Moreover, photo-oxidized milk exerted a pronounced detrimental impact on the glycerophospholipid metabolism of mice liver. The peroxisome proliferator-activated receptors (PPAR) signaling pathway enrichment appreciated in the animals that consumed photo-oxidized milk further supports the substantial negative influence of photo-oxidized milk on hepatic lipid metabolism. Gene set enrichment and interaction analyses revealed that photo-oxidized milk inhibited the cytochrome P450 pathway in mice, while also affecting other pathways associated with cellular stress response and lipid biosynthesis. CONCLUSION This comprehensive study provides significant evidence regarding the potential health risks associated with photo-oxidized milk, particularly in terms of hepatic oxidative damage. It establishes a scientific foundation for assessing the safety of such milk and ensuring the quality of dairy products.
Collapse
Affiliation(s)
- Sijia Tan
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, 100048, China; Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China
| | - Qiangqiang Li
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, China.
| | - Can Guo
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, China
| | - Sumeng Chen
- China Agricultural University, Beijing 100193, China
| | - Afaf Kamal-Eldin
- College of Food and Agriculture, Department of Food, Nutrition and Health (CFA), United Arab Emirates University, Al Ain 10008115551, United Arab Emirates
| | - Gang Chen
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, 100048, China.
| |
Collapse
|
17
|
Gonzalez-Orozco M, Tseng HC, Hage A, Xia H, Behera P, Afreen K, Peñaflor-Tellez Y, Giraldo MI, Huante M, Puebla-Clark L, van Tol S, Odle A, Crown M, Teruel N, Shelite TR, Menachery V, Endsley M, Endsley JJ, Najmanovich RJ, Bashton M, Stephens R, Shi PY, Xie X, Freiberg AN, Rajsbaum R. TRIM7 ubiquitinates SARS-CoV-2 membrane protein to limit apoptosis and viral replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599107. [PMID: 38948778 PMCID: PMC11212893 DOI: 10.1101/2024.06.17.599107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
SARS-CoV-2 is a highly transmissible virus that causes COVID-19 disease. Mechanisms of viral pathogenesis include excessive inflammation and viral-induced cell death, resulting in tissue damage. We identified the host E3-ubiquitin ligase TRIM7 as an inhibitor of apoptosis and SARS-CoV-2 replication via ubiquitination of the viral membrane (M) protein. Trim7 -/- mice exhibited increased pathology and virus titers associated with epithelial apoptosis and dysregulated immune responses. Mechanistically, TRIM7 ubiquitinates M on K14, which protects cells from cell death. Longitudinal SARS-CoV-2 sequence analysis from infected patients revealed that mutations on M-K14 appeared in circulating variants during the pandemic. The relevance of these mutations was tested in a mouse model. A recombinant M-K14/K15R virus showed reduced viral replication, consistent with the role of K15 in virus assembly, and increased levels of apoptosis associated with the loss of ubiquitination on K14. TRIM7 antiviral activity requires caspase-6 inhibition, linking apoptosis with viral replication and pathology.
Collapse
Affiliation(s)
- Maria Gonzalez-Orozco
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Hsiang-chi Tseng
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Adam Hage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Hongjie Xia
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX
| | - Padmanava Behera
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Kazi Afreen
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Yoatzin Peñaflor-Tellez
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Maria I. Giraldo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Matthew Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Lucinda Puebla-Clark
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| | - Sarah van Tol
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Abby Odle
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Matthew Crown
- Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Natalia Teruel
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Thomas R Shelite
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX
| | - Vineet Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Mark Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Janice J. Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
| | - Rafael J. Najmanovich
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Matthew Bashton
- Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX
| | | | - Ricardo Rajsbaum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ
| |
Collapse
|
18
|
Pfaffenbach M, Bolduc PN, Xin Z, Gao F, Evans R, Fang T, Chodaparambil JV, Henry KL, Li P, Mathieu S, Metrick C, Vera Rebollar JA, Gu RF, Mccarl CA, Silbereis J, Peterson EA. Discovery of BIO-8169─A Highly Potent, Selective, and Brain-Penetrant IRAK4 Inhibitor for the Treatment of Neuroinflammation. J Med Chem 2024; 67:8383-8395. [PMID: 38695469 DOI: 10.1021/acs.jmedchem.4c00560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Interleukin receptor associated kinase 4 (IRAK4) plays an important role in innate immune signaling through Toll-like and interleukin-1 receptors and represents an attractive target for the treatment of inflammatory diseases and cancer. We previously reported the development of a potent, selective, and brain-penetrant imidazopyrimidine series of IRAK4 inhibitors. However, lead molecule BIO-7488 (1) suffered from low solubility which led to variable PK, compound accumulation, and poor in vivo tolerability. Herein, we describe the discovery of a series of pyridone analogs with improved solubility which are highly potent, selective and demonstrate desirable PK profiles including good oral bioavailability and excellent brain penetration. BIO-8169 (2) reduced the in vivo production of pro-inflammatory cytokines, was well tolerated in safety studies in rodents and dog at margins well above the predicted efficacious exposure and showed promising results in a mouse model for multiple sclerosis.
Collapse
Affiliation(s)
- Magnus Pfaffenbach
- Department of Medicinal Chemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Philippe N Bolduc
- Department of Medicinal Chemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Zhili Xin
- Department of Medicinal Chemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Fang Gao
- Department of Medicinal Chemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ryan Evans
- Department of Medicinal Chemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Terry Fang
- Department of Acute Neurology, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Jayanth V Chodaparambil
- Physical Biochemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Kate L Henry
- Department of Acute Neurology, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Pei Li
- Drug Metabolism and Pharmacokinetics, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Steven Mathieu
- Pharmaceutical Operations & Technology, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Claire Metrick
- Physical Biochemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Jorge A Vera Rebollar
- Department of Multiple Sclerosis and Immunology, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Rong-Fang Gu
- Chemical Biology and Proteomics, Biogen Inc., Cambridge, Massachusetts 02142, United States
| | - Christie-Ann Mccarl
- Department of Multiple Sclerosis and Immunology, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - John Silbereis
- Department of Multiple Sclerosis and Immunology, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Emily A Peterson
- Department of Medicinal Chemistry, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
19
|
Groll T, Aupperle-Lellbach H, Mogler C, Steiger K. [Comparative pathology in oncology-Best practice]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:190-197. [PMID: 38602524 DOI: 10.1007/s00292-024-01327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Comparative experimental pathology is a research field at the interface of human and veterinary medicine. It is focused on the comparative study of similarities and differences between spontaneous and experimentally induced diseases in animals (animal models) compared to human diseases. The use of animal models for studying human diseases is an essential component of biomedical research. Interdisciplinary teams with species-specific expertise should collaborate wherever possible and maintain close communication. Mutual openness, cooperation, and willingness to learn form the basis for a fruitful collaboration. Research projects jointly led by or involving both animal and human pathologists make a significant contribution to high-quality biomedical research. Such approaches are promising not only in oncological research, as outlined in this article, but also in other research areas where animal models are regularly used, such as infectiology, neurology, and developmental biology.
Collapse
Affiliation(s)
- Tanja Groll
- Institut für Pathologie und Pathologische Anatomie, School of Medicine and Health, Technische Universität München, Trogerstraße 18, 81675, München, Deutschland
- Comparative Experimental Pathology (CEP), School of Medicine and Health, Technische Universität München, München, Deutschland
| | - Heike Aupperle-Lellbach
- Institut für Pathologie und Pathologische Anatomie, School of Medicine and Health, Technische Universität München, Trogerstraße 18, 81675, München, Deutschland
- Comparative Experimental Pathology (CEP), School of Medicine and Health, Technische Universität München, München, Deutschland
- LABOKLIN GmbH & Co. KG, Bad Kissingen, Deutschland
| | - Carolin Mogler
- Institut für Pathologie und Pathologische Anatomie, School of Medicine and Health, Technische Universität München, Trogerstraße 18, 81675, München, Deutschland
- Comparative Experimental Pathology (CEP), School of Medicine and Health, Technische Universität München, München, Deutschland
| | - Katja Steiger
- Institut für Pathologie und Pathologische Anatomie, School of Medicine and Health, Technische Universität München, Trogerstraße 18, 81675, München, Deutschland.
- Comparative Experimental Pathology (CEP), School of Medicine and Health, Technische Universität München, München, Deutschland.
| |
Collapse
|
20
|
Duan R, Huang K, Yu T, Chang C, Chu X, Huang Y, Zheng Z, Ma L, Li B, Yang T. Interleukin-2/anti-interleukin-2 complex attenuates inflammation in a mouse COPD model by expanding CD4 + CD25 + Foxp3 + regulatory T cells. Int Immunopharmacol 2024; 131:111849. [PMID: 38503017 DOI: 10.1016/j.intimp.2024.111849] [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: 10/29/2023] [Revised: 01/02/2024] [Accepted: 03/09/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND AND PURPOSE Chronic, nonspecific inflammation of the alveoli and airways is an important pathological feature of chronic obstructive pulmonary disease (COPD), while sustained inflammatory reactions can cause alveolar damage. Regulatory T cells (Tregs) inhibit inflammation, whereas the interleukin-2/anti-interleukin-2 complex (IL-2C) increases the number of Tregs; however, whether the IL-2C has a therapeutic role in COPD remains unknown. Therefore, this study investigated whether IL-2C alleviates lung inflammation in COPD by increasing the number of Tregs. EXPERIMENTAL APPROACH A mouse COPD model was created by exposing mice to lipopolysaccharides (LPS) and cigarette smoke (CS), and the effects of IL-2C treatment on COPD were evaluated. The number of Tregs in the spleen and lung, pulmonary pathological changes, and inflammatory damage were examined through flow cytometry, histopathology, and immunofluorescence, respectively. KEY RESULTS IL-2C increased the number of Treg cells in the spleen and lungs after exposure to CS and LPS, reduced the number of T helper 17 (Th17) cells in lung tissue, and improved the Th17/Treg balance. IL-2C decreased the number of inflammatory cells and reduced the levels of pro-inflammatory cytokines IL-6, TNF-α, IL-1β, CCL5, KC, and MCP-1 in bronchoalveolar lavage fluid and serum. IL-2C significantly reduced the pathological scores for lung inflammation, as well as decreased airway mucus secretion and infiltration of neutrophils and macrophages in the lungs. The depletion of Tregs using anti-CD25 antibodies eliminated the beneficial effects of IL-2C. CONCLUSIONS AND IMPLICATIONS IL-2C is a potential therapeutic agent for alleviating excessive inflammation in the lungs of patients with COPD.
Collapse
Affiliation(s)
- Ruirui Duan
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China; National Center for Respiratory Medicine, Beijing, China; State Key Laboratory of Respiratory Health and Multimorbidity, China
| | - Ke Huang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China; National Center for Respiratory Medicine, Beijing, China; State Key Laboratory of Respiratory Health and Multimorbidity, China
| | - Tao Yu
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chenli Chang
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xu Chu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China; National Center for Respiratory Medicine, Beijing, China; State Key Laboratory of Respiratory Health and Multimorbidity, China
| | - Yuhang Huang
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhoude Zheng
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Linxi Ma
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Baicun Li
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China; National Center for Respiratory Medicine, Beijing, China; State Key Laboratory of Respiratory Health and Multimorbidity, China.
| | - Ting Yang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China; National Center for Respiratory Medicine, Beijing, China; State Key Laboratory of Respiratory Health and Multimorbidity, China.
| |
Collapse
|
21
|
Mahmoodzadeh A, Valizadeh N, Edalati M, Khordadmehr M, Zakeri Z, Salehi R, Jarolmasjed S. Robust adhesive nanocomposite sponge composed of citric acid and nano clays modified cellulose for rapid hemostasis of lethal non-compressible hemorrhage. Carbohydr Polym 2024; 326:121614. [PMID: 38142075 DOI: 10.1016/j.carbpol.2023.121614] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/25/2023]
Abstract
Massive bleeding control plays the main role in saving people's lives in emergency situations. Herein, modified cellulose-based nanocomposite sponges by polydopamine (PDA) and laponite nano-clay was developed to sturdily deal with non-compressible lethal severe bleeding. PDA accomplishes supreme adhesion in the bleeding site (∼405 kPa) to form strong physical barrier and seal the position. Sponges super porous (∼70 % porosity) and super absorbent capacity (48 g blood absorbed per 1 g sponge) by concentrating the blood cells and platelets provides the requirements for primary hemostasis. Synergistically, the nanocomposite sponges' intelligent chemical structure induces hemostasis by activation of the XI, IX, X, II and FVII factors of intrinsic and extrinsic coagulation pathways. Excellent hemostatic performance of sponges in-vitro was assessed by RBC accumulation (∼100 %), blood clotting index (∼10 %), platelet aggregation/activation (∼93 %) and clotting time. The nanocomposite sponges depicted super performance in the fatal high-pressure non-compressible hemorrhage model by reducing of >2, 15 and 3 times in the bleeding amount at New Zealand rabbit's heart and liver, and rat's femoral artery bleeding models, respectively compared to commercial hemostatic agents (Pvalue˂0.001). The in-vivo host response results exhibited biosafety with no systemic and significant local inflammatory response by hematological, pathological and biochemical parameters assessments.
Collapse
Affiliation(s)
- Ahmad Mahmoodzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasrin Valizadeh
- Chemistry Department, Science Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mahdi Edalati
- Department of Laboratory Sciences, Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monireh Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ziba Zakeri
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Rumeli Feneri, 34450, Sariyer, Istanbul, Turkey
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyedhosein Jarolmasjed
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
22
|
Stokar-Regenscheit N, Bell L, Berridge B, Rudmann D, Tagle D, Hargrove-Grimes P, Schaudien D, Hahn K, Kühnlenz J, Ashton RS, Tseng M, Reichelt M, Laing ST, Kiyota T, Chamanza R, Sura R, Tomlinson L. Complex In Vitro Model Characterization for Context of Use in Toxicologic Pathology: Use Cases by Collaborative Teams of Biologists, Bioengineers, and Pathologists. Toxicol Pathol 2024; 52:123-137. [PMID: 38888280 DOI: 10.1177/01926233241253811] [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: 06/20/2024]
Abstract
Complex in vitro models (CIVMs) offer the potential to increase the clinical relevance of preclinical efficacy and toxicity assessments and reduce the reliance on animals in drug development. The European Society of Toxicologic Pathology (ESTP) and Society for Toxicologic Pathology (STP) are collaborating to highlight the role of pathologists in the development and use of CIVM. Pathologists are trained in comparative animal medicine which enhances their understanding of mechanisms of human and animal diseases, thus allowing them to bridge between animal models and humans. This skill set is important for CIVM development, validation, and data interpretation. Ideally, diverse teams of scientists, including engineers, biologists, pathologists, and others, should collaboratively develop and characterize novel CIVM, and collectively assess their precise use cases (context of use). Implementing a morphological CIVM evaluation should be essential in this process. This requires robust histological technique workflows, image analysis techniques, and needs correlation with translational biomarkers. In this review, we demonstrate how such tissue technologies and analytics support the development and use of CIVM for drug efficacy and safety evaluations. We encourage the scientific community to explore similar options for their projects and to engage with health authorities on the use of CIVM in benefit-risk assessment.
Collapse
Affiliation(s)
- Nadine Stokar-Regenscheit
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Luisa Bell
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | - Danilo Tagle
- National Center for Advancing Translational Sciences/National Institutes of Health, Bethesda, Maryland, USA
| | - Passley Hargrove-Grimes
- National Center for Advancing Translational Sciences/National Institutes of Health, Bethesda, Maryland, USA
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Kerstin Hahn
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Julia Kühnlenz
- Bayer SAS, CropScience, Pathology & Mechanistic Toxicology, Sophia Antipolis, France
| | - Randolph S Ashton
- University of Wisconsin-Madison, Madison, Wisconsin, USA
- Neurosetta LLC, Madison, Wisconsin, USA
| | - Min Tseng
- Genentech, South San Francisco, California, USA
| | | | | | | | | | | | - Lindsay Tomlinson
- Pfizer Inc., Drug Safety Research and Development, Cambridge, Massachusetts, USA
| |
Collapse
|
23
|
Bai J, Zhang C, Liu Y, Kuang N, Xu L, Xu Z, Wang H, Liu R. The therapeutic effect of Loranthus parasiticus lignan derivatives on collagen-induced arthritis in rats through the SHBG/NFκB pathway. Inflammopharmacology 2024; 32:873-883. [PMID: 38227094 DOI: 10.1007/s10787-023-01409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/10/2023] [Indexed: 01/17/2024]
Abstract
Lignan-rich beans, nuts, and various seeds are the main foods with antioxidative and hormone-modulating activities. Although the role of lignans in mediating hormone-dependent cancers and cardiovascular diseases is well characterized, the function of lignans in anti-arthritic activity and its underlying mechanisms remain unknown. Three new lignan derivatives, (-)-nortrachelogenin, trachelogenin, and matairesinol, were extracted from Loranthus parasiticus. After establishing the collagen-induced arthritis (CIA) model by intradermal injection of collagen, rats were treated with three new lignan derivatives ((-)-nortrachelogenin: 37%; trachelogenin: 27%; matairesinol: 25.7%) at a concentration of 50 mg/kg and 100 mg/kg, or methotrexate at 0.3 mg/kg. Mixed lignan derivatives significantly attenuated the immune responses in the joints of CIA rats, leading to lower levels of proinflammatory cytokines (IL-6 and TNF-α) and higher levels of free androgen in the serum compared to the CIA model. The results of molecular docking using AutoDock Vina showed that the lignan derivative (-)-nortrachelogenin was the most effective compound for binding to sex hormone-binding globulin (SHBG), thus inhibiting the activity of NFκB in LPS-stimulated macrophages. In this study, (-)-nortrachelogenin was identified as a novel natural lignan derivative with previously unrecognized anti-inflammatory activity. Its molecular mechanism appears related to the regulation of the NFκB/SHBG pathway. Our findings suggest that further application of sex hormone-like compounds in the treatment of rheumatoid arthritis and the potential clinical applications of (-)-nortrachelogenin are promising.
Collapse
Affiliation(s)
- Jiali Bai
- Clinical Pharmacy, Changzhi Maternal and Child Health Care Hospital, Changzhi, 046011, China
| | - Cong Zhang
- Department of Immunology, Medical College of Nanchang University, Nanchang, 330031, China
| | - Yulin Liu
- Department of Immunology, Medical College of Nanchang University, Nanchang, 330031, China
| | - Nanzhen Kuang
- Department of Immunology, Medical College of Nanchang University, Nanchang, 330031, China
| | - Liangquan Xu
- Department of Immunology, Medical College of Nanchang University, Nanchang, 330031, China
| | - Zhengang Xu
- Department of Immunology, Medical College of Nanchang University, Nanchang, 330031, China
| | - Haiwei Wang
- Obstetrics Department, Changzhi Maternal and Child Health Care Hospital, Changzhi, 046011, China
| | - Renping Liu
- Department of Immunology, Medical College of Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
24
|
Kvich L, Fritz BG, Zschach H, Terkelsen T, Raskov H, Høst-Rasmussen K, Jakobsen MR, Gheorghe AG, Gögenur I, Bjarnsholt T. Biofilms and core pathogens shape the tumor microenvironment and immune phenotype in colorectal cancer. Gut Microbes 2024; 16:2350156. [PMID: 38726597 PMCID: PMC11093030 DOI: 10.1080/19490976.2024.2350156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Extensive research has explored the role of gut microbiota in colorectal cancer (CRC). Nonetheless, metatranscriptomic studies investigating the in situ functional implications of host-microbe interactions in CRC are scarce. Therefore, we characterized the influence of CRC core pathogens and biofilms on the tumor microenvironment (TME) in 40 CRC, paired normal, and healthy tissue biopsies using fluorescence in situ hybridization (FISH) and dual-RNA sequencing. FISH revealed that Fusobacterium spp. was associated with increased bacterial biomass and inflammatory response in CRC samples. Dual-RNA sequencing demonstrated increased expression of pro-inflammatory cytokines, defensins, matrix-metalloproteases, and immunomodulatory factors in CRC samples with high bacterial activity. In addition, bacterial activity correlated with the infiltration of several immune cell subtypes, including M2 macrophages and regulatory T-cells in CRC samples. Specifically, Bacteroides fragilis and Fusobacterium nucleatum correlated with the infiltration of neutrophils and CD4+ T-cells, respectively. The collective bacterial activity/biomass appeared to exert a more significant influence on the TME than core pathogens, underscoring the intricate interplay between gut microbiota and CRC. These results emphasize how biofilms and core pathogens shape the immune phenotype and TME in CRC while highlighting the need to extend the bacterial scope beyond CRC pathogens to advance our understanding and identify treatment targets.
Collapse
Affiliation(s)
- Lasse Kvich
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Blaine Gabriel Fritz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Henrike Zschach
- Center for Health Data Science, University of Copenhagen, Copenhagen, Denmark
| | - Thilde Terkelsen
- Center for Health Data Science, University of Copenhagen, Copenhagen, Denmark
| | - Hans Raskov
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
| | - Kathrine Høst-Rasmussen
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
| | - Morten Ragn Jakobsen
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
| | - Alexandra Gabriella Gheorghe
- Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
25
|
Rais N, Ved A, Ahmad R, Parveen A. Research-based Analytical Procedures to Evaluate Diabetic Biomarkers and Related Parameters: In Vitro and In Vivo Methods. Curr Diabetes Rev 2024; 20:e201023222417. [PMID: 37867271 DOI: 10.2174/0115733998252495231011182012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/24/2023] [Accepted: 09/08/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND The degenerative tendency of diabetes leads to micro- and macrovascular complications due to abnormal levels of biochemicals, particularly in patients with poor diabetic control. Diabetes is supposed to be treated by reducing blood glucose levels, scavenging free radicals, and maintaining other relevant parameters close to normal ranges. In preclinical studies, numerous in vivo trials on animals as well as in vitro tests are used to assess the antidiabetic and antioxidant effects of the test substances. Since a substance that performs poorly in vitro won't perform better in vivo, the outcomes of in vitro studies can be utilized as a direct indicator of in vivo activities. OBJECTIVE The objective of the present study is to provide research scholars with a comprehensive overview of laboratory methods and procedures for a few selected diabetic biomarkers and related parameters. METHOD The search was conducted on scientific database portals such as ScienceDirect, PubMed, Google Scholar, BASE, DOAJ, etc. Conclusion: The development of new biomarkers is greatly facilitated by modern technology such as cell culture research, lipidomics study, microRNA biomarkers, machine learning techniques, and improved electron microscopies. These biomarkers do, however, have some usage restrictions. There is a critical need to find more accurate and sensitive biomarkers. With a few modifications, these biomarkers can be used with or even replace conventional markers of diabetes.
Collapse
Affiliation(s)
- Nadeem Rais
- Department of Pharmacy, Bhagwant University, Ajmer, Rajasthan 305004, India
| | - Akash Ved
- Goel Institute of Pharmaceutical Sciences, Lucknow, Uttar Pradesh 226028, India
| | - Rizwan Ahmad
- Department of Pharmacy, Vivek College of Technical Education, Bijnor, Uttar Pradesh 246701, India
| | - Aashna Parveen
- Faculty of Applied Science, Bhagwant Global University, Kotdwar, Uttarakhand 246149, India
| |
Collapse
|
26
|
Sepehri B, Darbani R, Mesgari-Abbasi M, Kheirouri S, Shanehbandi D, Khordadmehr M, Alizadeh M. The effects of short-time air pollution, SO2, and ozone on biochemical, histo-pathological, oxidative stress, and carcinogenesis related genes expressions in the liver of the rats. Hum Exp Toxicol 2024; 43:9603271241263569. [PMID: 39073095 DOI: 10.1177/09603271241263569] [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: 07/30/2024]
Abstract
OBJECTIVE OF THE RESEARCH Air pollution is a universal issue and has significant deleterious effects on both human health and also environment. The important indicators of air pollution include ozone (O3), particulate matter (PM), nitrogen dioxide (NO2), and sulfur dioxide (SO2). This research aims to investigate the impacts of ambient air pollution (AAP), SO2, and O3 on oxidative stress parameters, liver tissue histopathology, and expression of some carcinogenesis-related genes in the hepatic tissue of rats. MATERIALS AND METHODS 32 Wistar rats were randomly allocated to four groups: the control group, the AAP group, the SO2 group (10 ppm), and the ozone group (0.6 ppm). Over a period of five consecutive weeks, the rats were exposed to the specified pollutants for 3 h daily; liver tissues were harvested and instantly fixed with formalin. Pathological changes were assessed in the tissue samples. Additionally, the RT-qPCR technique was utilized to investigate Expression alterations of BAX, p-53, BCL2, caspase-3, caspase-8 and caspase-9. Furthermore, 30 milligrams of hepatic tissues were extracted to assess the activities of oxidative stress enzymes. RESULTS The liver catalase and MDA activity were elevated in the air pollution (p < .05). Also, liver GPx activity in air pollution and ozone groups was significant in comparison to the control group (p < .05). The SO2 group exhibited severe lesions in histopathology examinations. CONCLUSIONS The findings revealed an alteration in liver histopathology, an induction of oxidative stress, and the expression of some apoptosis-related genes in hepatic tissues after exposure to AAP, SO2, and O3.
Collapse
Affiliation(s)
- Bita Sepehri
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Darbani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sorayya Kheirouri
- Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology research center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monireh Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Mohammad Alizadeh
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Jarrell ZR, Lee CM, Kim KH, He X, Smith MR, Raha JR, Bhatnagar N, Orr M, Kang SM, Chen Y, Jones DP, Go YM. Metabolic reprograming and increased inflammation by cadmium exposure following early-life respiratory syncytial virus infection-the involvement of protein S-palmitoylation. Toxicol Sci 2023; 197:kfad112. [PMID: 37941452 PMCID: PMC10823773 DOI: 10.1093/toxsci/kfad112] [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] [Indexed: 11/10/2023] Open
Abstract
Early-life respiratory syncytial virus (RSV) infection (eRSV) is one of the leading causes of serious pulmonary disease in children. eRSV is associated with higher risk of developing asthma and compromised lung function later in life. Cadmium (Cd) is a toxic metal, widely present in the environment and in food. We recently showed that eRSV re-programs metabolism and potentiates Cd toxicity in the lung, and our transcriptome-metabolome-wide study showed strong associations between S-palmitoyl transferase expression and Cd-stimulated lung inflammation and fibrosis signaling. Limited information is available on the mechanism by which eRSV re-programs metabolism and potentiates Cd toxicity in the lung. In the current study, we used a mouse model to examine the role of protein S-palmitoylation (Pr-S-Pal) in low dose Cd-elevated lung metabolic disruption and inflammation following eRSV. Mice exposed to eRSV were later treated with Cd (3.3 mg CdCl2/L) in drinking water for 6 weeks (RSV+Cd). The role of Pr-S-Pal was studied using a palmitoyl transferase inhibitor, 2-bromopalmitate (BP, 10 µM). Inflammatory marker analysis showed that cytokines, chemokines and inflammatory cells were highest in the RSV+Cd group, and BP decreased inflammatory markers. Lung metabolomics analysis showed that pathways including phenylalanine, tyrosine and tryptophan, phosphatidylinositol and sphingolipid were altered across treatments. BP antagonized metabolic disruption of sphingolipid and glycosaminoglycan metabolism by RSV+Cd, consistent with BP effect on inflammatory markers. This study shows that Cd exposure following eRSV has a significant impact on subsequent inflammatory response and lung metabolism, which is mediated by Pr-S-Pal, and warrants future research for a therapeutic target.
Collapse
Affiliation(s)
- Zachery R Jarrell
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Choon-Myung Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, Georgia 30303, USA
| | - Xiaojia He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Matthew R Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia 30033, USA
| | - Jannatul R Raha
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, Georgia 30303, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, Georgia 30303, USA
| | - Michael Orr
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, Georgia 30303, USA
| | - Yan Chen
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
28
|
Ozan G, Cumbul A, Sumer E, Aydin A, Ekinci FY. Safety assessment of European cranberrybush (Viburnum opulus L.) fruit juice: Acute and subacute oral toxicity. Food Chem Toxicol 2023; 181:114082. [PMID: 37783419 DOI: 10.1016/j.fct.2023.114082] [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: 07/22/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
European cranberrybush (ECB) (Viburnum opulus L.) fruits are abundant in phenolic compounds associated with various health benefits. However, the toxicity and safety of ECB juice have not been systematically studied. In the present study, acute and subacute oral toxicities of ECB fruit juice were evaluated on Sprague-Dawley rats and BALB/c mice to establish a toxicity profile. In acute tests, a single administration of 2000 mg/kg body weight of extract to rats exhibited no clinical signs of toxicity or mortality, indicating that the lethal dose (LD50) was over 2000 mg/kg. In subacute tests, repeated administration for 28 days at 0 (control), 500, and 2000 mg/kg doses of extract in mice did not display adverse clinical signs or deaths. However, in the 2000 mg/kg subacute group, platelet counts were significantly high, which correlated with histopathological analyses revealing that ECB extract at 2000 mg/kg was toxic to the kidney, liver, and adipose tissue. The NOAEL value of ECB extract was found as 500 mg/kg/day, but further sub-chronic and chronic toxicity studies are warranted to comprehensively evaluate the long-term safety implications. The study's results emphasize the importance of considering the dosage of dietary supplements containing high levels of phenolic compounds over an extended period to avoid potential cumulative effects from prolonged consumption of high doses.
Collapse
Affiliation(s)
- Gizem Ozan
- Yeditepe University, Faculty of Engineering, Department of Food Engineering, Istanbul, Turkiye; Yeditepe University, Graduate School of Natural and Applied Sciences, Biotechnology Program, Istanbul, Turkiye
| | - Alev Cumbul
- Yeditepe University, Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkiye
| | - Engin Sumer
- Yeditepe University, Faculty of Medicine, Experimental Research Center, Istanbul, Turkiye
| | - Ahmet Aydin
- Yeditepe University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Istanbul, Turkiye
| | - F Yesim Ekinci
- Yeditepe University, Faculty of Engineering, Department of Food Engineering, Istanbul, Turkiye; Yeditepe University, Graduate School of Natural and Applied Sciences, Biotechnology Program, Istanbul, Turkiye.
| |
Collapse
|
29
|
Cao Y, Tao F, Yu Y, Song L, Zhang R, Feng J, Zhai Q, Xue P. Safety evaluation of rare ginsenosides of stems and leaves from American ginseng: 90-day exposure toxicity study combined with intestinal flora analysis and metabonomics in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115429. [PMID: 37660532 DOI: 10.1016/j.ecoenv.2023.115429] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/24/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Rare ginsenosides have already been widely applied in many fields, including health food and bio-medicine. The human being can expose to rare ginsenosides directly or indirectly increasingly. However, there are few studies on the safety assessment of rare ginsenoside mixtures. In the present study, the sub-chronic toxicity of rare ginsenosides for 90 days on SD rats was performed by combining the intestinal flora analysis and urine metabonomics aiming to illustrate the safety of long-term consumption of rare ginsenosides and the potential damage for liver and intestinal. 48 adult rats were divided into four groups: control (0 mg/kg), low-dose (60 mg/kg), medium-dose (200 mg/kg), and high-dose (600 mg/kg). Rats in the high-dose group showed inflammatory changes in their livers and intestines. The strong bactericidal effect of rare ginsenosides caused intestinal flora disorder and changed the structure of intestinal flora in rats, thus inducing intestinal damage in rats. In the high-dose group, levels of alanine aminotransferase (ALT), lactate dehydrogenase (LDH), and alkaline phosphatase (AKP) increased significantly. As a result of the high-dose treatment, certain metabolic pathways were altered, such as vitamin B6 metabolism, methionine metabolism, glutathione metabolism, and others. These results indicated that high doses of rare ginsenosides induced liver injury by affecting the above metabolic pathways. Rare ginsenosides with no observed adverse effect level (NOAEL) were below 200 mg/kg/day in vivo. Thus, this present study provides insight into the rational use of rare ginsenosides.
Collapse
Affiliation(s)
- Yuqing Cao
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Feiyan Tao
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Yuan Yu
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Linmeng Song
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Ruoyu Zhang
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Jing Feng
- School of Rehabilitation, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Qingfeng Zhai
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China.
| | - Peng Xue
- School of Public Health, Weifang Medical University, Weifang 261053, People's Republic of China.
| |
Collapse
|
30
|
Ghiasi F, Mesgari-Abbasi M, Khordadmehr M, Imani S, Hosseinzadeh F. Chronic Kombucha Beverage Consumption Attenuates Inflammatory Markers and Histopathology of Brain Tissue in Transnet Global Brain Ischemia in Rats. Neurochem Res 2023; 48:3202-3211. [PMID: 37402035 DOI: 10.1007/s11064-023-03980-2] [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: 08/27/2022] [Revised: 05/22/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023]
Abstract
There is evidence that kombucha beverage (KB), a traditional fermented beverage, has a preventive effect on experimental brain ischemia. According to our previous studies, pre-treatment of KB attenuates brain edema and improves motor skills and oxidative stress in a rat model of global brain ischemia. This study was designed to evaluate the effects of the pre-treatment of KB, as a novel agent, on pro-inflammatory parameters and brain histopathology changes following global brain ischemia. Adult male Wistar rats were randomly divided into the sham, the control, and the groups treated with kombucha (KB1 and KB2 groups). KB at doses 1 and 2 mL/kg was prescribed two-week consecutive days before induction of global brain ischemia. Global brain ischemia was induced by blocking common carotid arteries for 60 min and the following reperfusion by 24 h. The serum and brain levels of tumor necrosis factor-α(TNF-α), IL-1β, histopathological change, and infarct volume are determined using the ELISA, hematoxylin and eosin (H&E), and 2,3,5-triphenyl tetrazolium chloride (TTC) staining, respectively. This study indicated that pre-treatment of KB significantly reduced infarct volume, the serum, and brain levels of TNF-α and IL-1β. The histopathological finding of the brain tissue confirmed a protective role for pre-treatment KB in the ischemic rats. Thus, the present study showed that the beneficial effects of KB pre-treatment on brain ischemic may be mediated by decreasing pro-inflammatory parameters.
Collapse
Affiliation(s)
- Fariba Ghiasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Monireh Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Sepideh Imani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fezzeh Hosseinzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Physiology, Sarab Faculty of Medicine Sciences, Sarab, Iran.
| |
Collapse
|
31
|
Owumi SE, Umez AO, Arunsi U, Irozuru CE. Dietary aflatoxin B1 and antimalarial-a lumefantrine/artesunate-therapy perturbs male rat reproductive function via pro-inflammatory and oxidative mechanisms. Sci Rep 2023; 13:12172. [PMID: 37500724 PMCID: PMC10374580 DOI: 10.1038/s41598-023-39455-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/13/2023] [Indexed: 07/29/2023] Open
Abstract
We investigated the impact of Coartem™ (COA) and aflatoxin B1 (AFB1) on rats' hypothalamus, epididymis, and testis. Male rats were randomly grouped (n = 5 rats) and treated: control group (corn oil), AFB1 (70 µg/kg), COA (5 mg/kg), COA + AFB1 (5 + 0.035 mg/kg) and COA + AFB1 (5 + 0.07 mg/kg) for 28 days. Blood samples were collected for serum prolactin, testosterone, follicle-stimulating and luteinising hormones (FSH and LH) assay upon sacrifice. The semen, hypothalamus, epididymis, and testes were harvested for morphological, biochemical, and histopathology determination of oxidative, inflammation stress, genomic integrity, and pathological alterations. Exposure to the COA and AFB1 caused the cauda epididymal spermatozoa to display low motility, viability, and volume, with increased abnormalities. Hormonal disruption ensued in animals exposed to COA and AFB1 alone or together, exemplified by increased prolactin, and decreased testosterone, FSH and LH levels. Treatment-related reduction in biomarkers of testicular metabolism-acid and alkaline phosphatases, glucose-6-phosphate dehydrogenase, and lactate dehydrogenase-were observed. Also, COA and AFB1 treatment caused reductions in antioxidant (Glutathione and total thiols) levels and antioxidant enzyme (Catalase, superoxide dismutase, glutathione peroxidase, and glutathione-S-transferase) activities in the examined organs. At the same time, treatment-related increases in DNA damage (p53), oxidative stress (xanthine oxidase, reactive oxygen and nitrogen species and lipid peroxidation), inflammation (nitric oxide and tumour necrosis factor-alpha), and apoptosis (caspase-9, and -3) were observed. Chronic exposure to COA and AFB1 led to oxidative stress, inflammation, and DNA damage in male rats' hypothalamic-reproductive axis, which might potentiate infertility if not contained.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, 200004, Nigeria.
- ChangeLab-changing Lives, Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, University of Ibadan, Rm NB 302, Ibadan, 200005, Oyo State, Nigeria.
| | - Angel O Umez
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, 200004, Nigeria
| | - Uche Arunsi
- School of Chemistry and Biochemistry, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0400, USA
| | - Chioma E Irozuru
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| |
Collapse
|
32
|
Development and Validation of a Multiparametric Semiquantitative Scoring System for the Histopathological Assessment of Ischaemia Severity in Skeletal Muscle. J Tissue Eng Regen Med 2023. [DOI: 10.1155/2023/5592455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Skeletal muscle is one of the most abundant and dynamic tissues of the body, with a strong regenerative capacity. Muscle injuries can occur as a result of a variety of events, including tissue ischaemia. Lower limb ischaemia occurs when there is an insufficient nutrient and oxygen supply, often caused by stenosis of the arteries due to atherosclerosis. The aim of this study was to develop and validate a multiparametric scoring tool for assessing ischaemia severity in skeletal muscle in a commonly used preclinical animal model. Tissue ischaemia was surgically induced in mice by ligation and excision of the femoral artery. Calf muscles were carefully dissected, prepared for histological analysis, and scored for inflammation, fibrosis, necrosis, adipocyte infiltration, and muscle fibre degeneration/regeneration. Kendall’s coefficient of concordance (W) showed a very good agreement between the appraisers when scoring each individual histological feature: inflammation (W = 0.92,
), fibrosis (W = 0.94,
), necrosis (W = 0.77,
), adipocyte infiltration (W = 0.91,
), and fibre degeneration/regeneration (W = 0.86,
). Intrarater agreement was also excellent (W = 0.94 or more,
). There was a statistically significant negative association between the level of muscle ischaemia damage and the calf muscle weight and skeletal muscle fibre diameter. Here, we have developed and validated a new multiparametric, semiquantitative scoring system for assessing skeletal muscle damage due to ischaemia, with excellent inter- and intrarater reproducibility. This scoring system can be used for assessing treatment efficacy in preclinical models of hind limb ischaemia.
Collapse
|
33
|
Cruces P, Erranz B, González C, Diaz F. Morphological Differences between Patient Self-inflicted and Ventilator-induced Lung Injury: An Experimental Study. Am J Respir Crit Care Med 2023; 207:780-783. [PMID: 36450096 PMCID: PMC10037468 DOI: 10.1164/rccm.202207-1360le] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Pablo Cruces
- Centro de Investigación de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
- Unidad de Paciente Crítico Pediátrico, Hospital El Carmen Dr. Luis Valentín Ferrada, Santiago, Chile
| | - Benjamín Erranz
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile; and
| | - Carlos González
- Centro de Investigación de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Franco Diaz
- Unidad de Paciente Crítico Pediátrico, Hospital El Carmen Dr. Luis Valentín Ferrada, Santiago, Chile
- Unidad de Investigación y epidemiología clínica, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| |
Collapse
|
34
|
D’Amico R, Tomasello M, Impellizzeri D, Cordaro M, Siracusa R, Interdonato L, Abdelhameed AS, Fusco R, Calabrese V, Cuzzocrea S, Di Paola R. Mechanism of Action of Natural Compounds in Peripheral Multiorgan Dysfunction and Hippocampal Neuroinflammation Induced by Sepsis. Antioxidants (Basel) 2023; 12:antiox12030635. [PMID: 36978883 PMCID: PMC10045853 DOI: 10.3390/antiox12030635] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Bacterial sepsis induces the production of excessive pro-inflammatory cytokines and oxidative stress, resulting in tissue injury and hyperinflammation. Patients recovering from sepsis have increased rates of central nervous system (CNS) morbidities, which are linked to long-term cognitive impairment, such as neurodegenerative pathologies. This paper focuses on the tissue injury and hyperinflammation observed in the acute phase of sepsis and on the development of long-term neuroinflammation associated with septicemia. Here we evaluate the effects of Coriolus versicolor administration as a novel approach to treat polymicrobial sepsis. Rats underwent cecal ligation and perforation (CLP), and Coriolus versicolor (200 mg/kg in saline) was administered daily by gavage. Survival was monitored, and tissues from vital organs that easily succumb to infection were harvested after 72 h to evaluate the histological changes. Twenty-eight days after CLP, behavioral analyses were performed, and serum and brain (hippocampus) samples were harvested at four weeks from surgery. Coriolus versicolor increased survival and reduced acute tissue injury. Indeed, it reduced the release of pro-inflammatory cytokines in the bloodstream, leading to a reduced chronic inflammation. In the hippocampus, Coriolus versicolor administration restored tight junction expressions, reduce cytokines accumulation and glia activation. It also reduced toll-like receptor 4 (TLR4) and neuronal nitric oxide synthase (nNOS) and the NLR family pyrin domain containing 3 (NLRP3) inflammasome components expression. Coriolus versicolor showed antioxidant activities, restoring glutathione (GSH) levels and catalase and superoxide dismutase (SOD) activities and reducing lipid peroxidation, nitrite and reactive oxygen species (ROS) levels. Importantly, Coriolus versicolor reduced amyloid precursor protein (APP), phosphorylated-Tau (p-Tau), pathologically phosphorylated tau (PHF1), phosphorylated tau (Ser202 and Thr205) (AT8), interferon-induced transmembrane protein 3 (IFITM3) expression, and β-amyloid accumulation induced by CLP. Indeed, Coriolus versicolor restored synaptic dysfunction and behavioral alterations. This research shows the effects of Coriolus versicolor administration on the long-term development of neuroinflammation and brain dysfunction induced by sepsis. Overall, our results demonstrated that Coriolus versicolor administration was able to counteract the degenerative process triggered by sepsis.
Collapse
Affiliation(s)
- Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Mario Tomasello
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Ali Saber Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 14451, Saudi Arabia
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
- Correspondence:
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Vererinary Sciences, University of Messina, Viale Annunziata, 98168 Messina, Italy
| |
Collapse
|
35
|
Munipalli SB, Yenugu S. Uroplakin 1a Knockout Mice Display Marginal Reduction in Fecundity, Decreased Bacterial Clearance Capacity, and Drastic Changes in the Testicular Transcriptome. Reprod Sci 2023; 30:914-927. [PMID: 36042152 DOI: 10.1007/s43032-022-01057-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/02/2022] [Indexed: 10/14/2022]
Abstract
Uroplakins (UPKs) form physical and chemical barriers in the bladder and other urinary tract tissues. We previously reported the identification and localization of UPKs in the male reproductive tract of rat. In this study, we characterized Upk1a knockout mice and report a marginal reduction in fecundity associated with significant decrease in sperm count. Upk1a mice had lower bacterial clearance capacity when challenged with uropathogenic Escherichia coli for 1 to 5 days. High-throughput analyses of testicular transcriptome indicated that 1128 genes that are expressed in testis of wild-type mice were completely absent in the knockout, while 2330 genes were found to be expressed only in the testis of knockout mice. Furthermore, differential regulation of 148 (67 upregulated and 81 downregulated) was observed. Gene ontology analyses indicated that processes related to integral components of membrane (plasma membrane), G-protein receptor activity and signaling, olfactory receptor activity and perception of smell, organization of extracellular space/region, immune and inflammatory responses to pathogens, spermatid development, meiotic cell cycle, and formation of synaptonemal complex were affected. Results of this study provide evidence on the possible multi-functional role of Upk1a in male reproductive tract and in other tissues as well.
Collapse
Affiliation(s)
| | - Suresh Yenugu
- Department of Animal Biology, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
36
|
Han M, Jeong S, Song J, Park SJ, Min Lee C, Lee K, Park SM. Association between the dual use of electronic and conventional cigarettes and NAFLD status in Korean men. Tob Induc Dis 2023; 21:31. [PMID: 36844383 PMCID: PMC9951190 DOI: 10.18332/tid/159167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 02/26/2023] Open
Abstract
INTRODUCTION This study investigated the association between smoking types, including dual use (usage of both combustible cigarettes and e-cigarettes), and non-alcoholic fatty liver disease (NAFLD) status in Korean men. METHODS Data from the 7th and 8th Korea National Health and Nutrition Examination Survey (KNHANES) 2016-2020 were used. The presence of NAFLD was defined by the respective cut-off values for the Hepatic Steatosis Index (HSI), NAFLD Ridge Score (NRS), and Korea National Health and Nutrition Examination Survey NAFLD score (KNS). Multivariate logistic regression analyses were used to determine the associations between smoking types and NAFLD as determined by HSI, NRS, and KNS. RESULTS After adjustment for confounders, an independent association was observed between dual use and NAFLD (HSI: AOR=1.47; 95% CI: 1.08-1.99, p=0.014; NRS: AOR=2.21; 95% CI: 1.70-2.86, p=0.000; KNS: AOR=1.35; 95% CI: 1.01-1.81, p=0.045). Cigarette only smokers also had significantly higher odds of NAFLD compared to never smokers for all of the NAFLD indices (HSI: AOR=1.22; 95% CI: 1.05-1.42, p=0.008; NRS: AOR=2.13; 95% CI: 1.87-2.42, p=0.000; KNS: AOR=1.33; 95% CI: 1.14-1.55, p=0.000). In subgroup analyses, no significant interaction effects were found for age, BMI, alcohol consumption, income, physical activity, and the diagnosis of T2DM. Moreover, cigarette only smokers and dual users differed significantly in terms of log-transformed urine cotinine and pack-years. The relationship between smoking type and pack-years was attenuated after stratification by age. CONCLUSIONS This study shows that the dual use of e-cigarettes and combustible cigarettes is associated with NAFLD. Age differences may explain why dual users, with a greater proportion of young people, appear to have fewer pack-years than cigarette only smokers. Further research should be conducted to investigate the adverse effects of dual use on hepatic steatosis.
Collapse
Affiliation(s)
- Minjung Han
- Department of Family Medicine, Myongji Hospital, Goyang, Republic of Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam, Republic of Korea,Institute for Biomedical Informatics, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Jihun Song
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Sun Jae Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Cheol Min Lee
- Department of Family Medicine, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea,Department of Family Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kiheon Lee
- Department of Family Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Family Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea,Department of Family Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Family Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
37
|
Ceran F, Pilanci O, Ozel A, Ilbay G, Karabacak R, Kanter M, Ilbay K, Kuvat SV. Use of acellular dermal matrix in peripheral nerve reconstruction: an experimental study on rat sciatic nerve defect. J Plast Surg Hand Surg 2023; 57:445-452. [PMID: 36476277 DOI: 10.1080/2000656x.2022.2152824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND In patients with nerve tissue defects, the use of autologous nerve grafts is the standard method of treatment. Alternatives to autologous, nerve grafts have attracted the attention of reconstructive surgeons. In this study, the results of nerve repairs using acellular dermal matrix (ADM) in an experimental rat sciatic nerve defect model are presented. METHODS Thirty-six Sprague-Dawley rats were randomized into 5 groups: Group 1: control group, Group 2: negative control group (n = 6), Group 3: autologous nerve graft group (n = 10), Group 4: donor site entubulated with ADM group (n = 10); and Group 5: nerve graft entubulated with ADM group (n = 10). The animals in each group were evaluated for electrophysiologic functions, gastrocnemius muscle weight and histomorphology on the 3rd and 6th month. RESULTS The compound muscle action potential was observed to be distinctly lower in Groups 3, 4 and 5 in comparison to the control group. In Group 4, the gastrocnemius ratio (GCR) values on the 6th month were statistically significantly lower than the GCR values in Group 3 and Group 5, The histological scores and myelinated axonal counts in Group 5 were statistically significantly higher than the values in Group 3 and Group 4. CONCLUSION The results of this study showed that wrapping ADM around nerve grafts resulted in better outcomes with respect to nerve healing.
Collapse
Affiliation(s)
- Fatih Ceran
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medicalpark Hospital, Batman, Turkey
| | - Ozgur Pilanci
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Asuman Ozel
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Gul Ilbay
- Faculty of Medicine, Department of Physiology, Kocaeli University, Kocaeli, Turkey
| | - Rukiye Karabacak
- Faculty of Medicine, Department of Histology, Medeniyet University, Istanbul, Turkey
| | - Mehmet Kanter
- Faculty of Medicine, Department of Histology, Medeniyet University, Istanbul, Turkey
| | - Konuralp Ilbay
- Faculty of Medicine, Department of Neurosurgery, Kocaeli University, Kocaeli, Turkey
| | - Samet Vasfi Kuvat
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
38
|
Le-Trilling VTK, Ebel JF, Baier F, Wohlgemuth K, Pfeifer KR, Mookhoek A, Krebs P, Determann M, Katschinski B, Adamczyk A, Lange E, Klopfleisch R, Lange CM, Sokolova V, Trilling M, Westendorf AM. Acute cytomegalovirus infection modulates the intestinal microbiota and targets intestinal epithelial cells. Eur J Immunol 2023; 53:e2249940. [PMID: 36250419 DOI: 10.1002/eji.202249940] [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: 04/12/2022] [Revised: 08/29/2022] [Accepted: 10/14/2022] [Indexed: 02/04/2023]
Abstract
Primary and recurrent cytomegalovirus (CMV) infections frequently cause CMV colitis in immunocompromised as well as inflammatory bowel disease (IBD) patients. Additionally, colitis occasionally occurs upon primary CMV infection in patients who are apparently immunocompetent. In both cases, the underlying pathophysiologic mechanisms are largely elusive - in part due to the lack of adequate access to specimens. We employed the mouse cytomegalovirus (MCMV) model to assess the association between CMV and colitis. During acute primary MCMV infection of immunocompetent mice, the gut microbial composition was affected as manifested by an altered ratio of the Firmicutes to Bacteroidetes phyla. Interestingly, these microbial changes coincided with high-titer MCMV replication in the colon, crypt hyperplasia, increased colonic pro-inflammatory cytokine levels, and a transient increase in the expression of the antimicrobial protein Regenerating islet-derived protein 3 gamma (Reg3γ). Further analyses revealed that murine and human intestinal epithelial cell lines, as well as primary intestinal crypt cells and organoids represent direct targets of CMV infection causing increased cell death. Accordingly, in vivo MCMV infection disrupted the intestinal epithelial barrier and increased apoptosis of intestinal epithelial cells. In summary, our data show that CMV transiently induces colitis in immunocompetent hosts by altering the intestinal homeostasis.
Collapse
Affiliation(s)
| | - Jana-Fabienne Ebel
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Franziska Baier
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kerstin Wohlgemuth
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kai Robin Pfeifer
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Aart Mookhoek
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Madita Determann
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Benjamin Katschinski
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Erik Lange
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Free University of Berlin, Berlin, Germany
| | - Christian M Lange
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Viktoriya Sokolova
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
39
|
He Q, Wu J, Ke J, Zhang Q, Zeng W, Luo Z, Gong J, Chen Y, He Z, Lan P. Therapeutic role of ursodeoxycholic acid in colitis-associated cancer via gut microbiota modulation. Mol Ther 2023; 31:585-598. [PMID: 38556635 PMCID: PMC9931610 DOI: 10.1016/j.ymthe.2022.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/21/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a predisposing factor for colitis-associated cancer (CAC). The association between bile acids and the gut microbiota has been demonstrated in colon neoplasia; however, the effect of ursodeoxycholic acid (UDCA) on gut microbiota alteration in development of colitis and CAC is unknown. Our analysis of publicly available datasets demonstrated the association of UDCA treatment and accumulation of Akkermansia. UDCA-mediated alleviation of DSS-induced colitis was microbially dependent. UDCA treatment significantly upregulated Akkermansia colonization in a mouse model. Colonization of Akkermansia was associated with enhancement of the mucus layer upon UDCA treatment as well as activation of bile acid receptors in macrophages. UDCA played a role in CAC prevention and treatment in the AOM-DSS and ApcMin/+-DSS models through downregulation of inflammation and accumulation of Akkermansia. This study suggests that UDCA intervention could reshape intestinal gut homeostasis, facilitating colonization of Akkermansia and preventing and treating colitis and CAC.
Collapse
Affiliation(s)
- Qilang He
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Jinjie Wu
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Jia Ke
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Qiang Zhang
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Wanyi Zeng
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Zhanhao Luo
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Junli Gong
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China
| | - Yuan Chen
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China
| | - Zhen He
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China.
| | - Ping Lan
- The Sixth Affiliated Hospital, School of Medicine, Sun Yat-sen University, Guangzhou, 510655 Guangdong, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655 Guangdong, China.
| |
Collapse
|
40
|
Owumi SE, Otunla MT, Elerewe OO, Arunsi UO. Co-exposure to aflatoxin B1 and therapeutic coartem worsens hepatic and renal function through enhanced oxido-inflammatory responses and apoptosis in rats. Toxicon 2023; 222:106988. [PMID: 36473514 DOI: 10.1016/j.toxicon.2022.106988] [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: 07/21/2022] [Revised: 11/14/2022] [Accepted: 11/30/2022] [Indexed: 12/09/2022]
Abstract
Aflatoxin B1 (AFB1) is a mycotoxin synthesised as a secondary metabolite by members of the Aspergillus species contaminating agricultural produce. Aspergillus species thrive in tropical climes, endemic to malaria. Artemisinin-based combination therapies (ACTs) effectively treat and prevent malaria recrudescence; Coartem (COA) is an ACT whose toxicity is evident. Although there are scanty studies on COA toxicity, the scientific literature is replete on AFB1 toxic effects -including carcinogenicity. The current research investigates AFB1 and COA toxicity in experimental Wistar rats' hepatorenal systems. Thirty albino rats were randomly grouped into five cohorts (n = 6) and treated as follows: Group I: Untreated control (2 mL/kg of corn oil); group II: AFB1 alone (70 μg/kg); group III: COA alone (5 mg/kg); group IV: COA and a low dose of AFB11 (5 mg/kg & 35 μg/kg); while Group V: COA and a high dose AFB12 (5 mg/kg & 70 μg/kg) by gavage. Our results show that exposure to AFB1 and COA significantly (p < 0.05) reduced superoxide dismutase, catalase, glutathione peroxidase, and glutathione-S-transferase activities, besides reduced glutathione and total sulfhydryl groups level. Reactive oxygen and nitrogen species, lipid peroxidation, 8-hydroxy-2'-deoxyguanosine, nitric oxide, xanthine oxidase, and myeloperoxidase levels were increased (p < 0.05) in rats co-treated with COA and AFB1. Cell death was aggravated in COA and AFB1 groups, exemplified by increased Caspase-3 and 9 activities and alterations in the typical histological features of experimental rats' livers and kidneys. Finally, rats co-treated with AFB1 and COA experienced increased hepatorenal dysregulation, oxidative and inflammatory tissue damage, and apoptotic cell death. All the observed systemic perturbations occurred dose-dependently. It is crucial, therefore, to prevent AFB1 dietary contaminations during COA therapeutic regimen due to increased pathophysiological damage exerted on experimental rat liver and kidneys, as evidenced in this study.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, 200004, Nigeria.
| | - Moses T Otunla
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, 200004, Nigeria
| | - Oyindamola O Elerewe
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, 200004, Nigeria
| | - Uche O Arunsi
- Department of Cancer Immunology and Biotechnology, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
41
|
Noh H, Yoon S, Kim SH, Kim J, Seo JS, Kim JJ, Park IH, Oh J, Bae JY, Lee GE, Woo SJ, Seo SM, Kim NW, Lee YW, Jang HJ, Hong SM, An SH, Lyoo KS, Yeom M, Lee H, Jung B, Yoon SW, Kang JA, Seok SH, Lee YJ, Kim SY, Kim YB, Hwang JY, On D, Lim SY, Kim SP, Jang JY, Lee H, Kim K, Lee HJ, Kim HB, Kim SB, Park JW, Jeong DG, Song D, Choi KS, Lee HY, Choi YK, Choi JA, Song M, Park MS, Seo JY, Shin JS, Yun JW, Nam KT, Seong JK. Establishment of multicenter COVID-19 therapeutics preclinical test system in Republic of Korea. Pulm Pharmacol Ther 2023; 80:102189. [PMID: 36634813 PMCID: PMC9829441 DOI: 10.1016/j.pupt.2023.102189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/03/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Throughout the recent COVID-19 pandemic, South Korea led national efforts to develop vaccines and therapeutics for SARS-CoV-2. The project proceeded as follows: 1) evaluation system setup (including Animal Biosafety Level 3 (ABSL3) facility alliance, standardized nonclinical evaluation protocol, and laboratory information management system), 2) application (including committee review and selection), and 3) evaluation (including expert judgment and reporting). After receiving 101 applications, the selection committee reviewed pharmacokinetics, toxicity, and efficacy data and selected 32 final candidates. In the nonclinical efficacy test, we used golden Syrian hamsters and human angiotensin-converting enzyme 2 transgenic mice under a cytokeratin 18 promoter to evaluate mortality, clinical signs, body weight, viral titer, neutralizing antibody presence, and histopathology. These data indicated eight new drugs and one repositioned drug having significant efficacy for COVID-19. Three vaccine and four antiviral drugs exerted significant protective activities against SARS-CoV-2 pathogenesis. Additionally, two anti-inflammatory drugs showed therapeutic effects on lung lesions and weight loss through their mechanism of action but did not affect viral replication. Along with systematic verification of COVID-19 animal models through large-scale studies, our findings suggest that ABSL3 multicenter alliance and nonclinical evaluation protocol standardization can promote reliable efficacy testing against COVID-19, thus expediting medical product development.
Collapse
Affiliation(s)
- Hyuna Noh
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suhyeon Yoon
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung-Hee Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jiseon Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jung Seon Seo
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jeong Jin Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - In Ho Park
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea,Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jooyeon Oh
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, Biosafety Center, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Gee Eun Lee
- Department of Microbiology, Institute for Viral Diseases, Biosafety Center, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sun-Je Woo
- Science Unit, International Vaccine Institute, Seoul, 08826, Republic of Korea
| | - Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Na-Won Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Youn Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 13488, Republic of Korea
| | - Hui Jeong Jang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 13488, Republic of Korea
| | - Seung-Min Hong
- Laboratory of Avian Diseases, BK21 plus Program for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Se-Hee An
- Laboratory of Avian Diseases, BK21 plus Program for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwang-Soo Lyoo
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, 54531, Republic of Korea
| | - Minjoo Yeom
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Hanbyeul Lee
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Bud Jung
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Sun-Woo Yoon
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jung-Ah Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, 24341, Republic of Korea
| | - Yu Jin Lee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, 24341, Republic of Korea
| | - Seo Yeon Kim
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam, 13488, Republic of Korea
| | - Young Been Kim
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam, 13488, Republic of Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam, 13488, Republic of Korea
| | - Dain On
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea,Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Soo-Yeon Lim
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sol Pin Kim
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Yun Jang
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea,College of Pharmacy, Dongguk University, Seoul, 04620, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Kyoungmi Kim
- Department of Biomedical Sciences and Department of Physiology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Hyo-Jung Lee
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Hong Bin Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620, Republic of Korea
| | - Sun Bean Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, 24341, Republic of Korea
| | - Dae Gwin Jeong
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Daesub Song
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Kang-Seuk Choi
- Laboratory of Avian Diseases, BK21 plus Program for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 13488, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jung-ah Choi
- Science Unit, International Vaccine Institute, Seoul, 08826, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul, 08826, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Biosafety Center, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jun-Young Seo
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jeon-Soo Shin
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea,Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea,Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea,Corresponding author. Laboratory of Veterinary Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea,Corresponding author
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea,Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, 08826, Republic of Korea,Corresponding author. Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
42
|
Amni F, Maleki-Ravasan N, Nateghi-Rostami M, Hadighi R, Karimian F, Meamar AR, Badirzadeh A, Parvizi P. Co-infection of Phlebotomus papatasi (Diptera: Psychodidae) gut bacteria with Leishmania major exacerbates the pathological responses of BALB/c mice. Front Cell Infect Microbiol 2023; 13:1115542. [PMID: 36779192 PMCID: PMC9909354 DOI: 10.3389/fcimb.2023.1115542] [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: 12/04/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Clinical features and severity of the leishmaniasis is extremely intricate and depend on several factors, especially sand fly-derived products. Bacteria in the sand fly's gut are a perpetual companion of Leishmania parasites. However, consequences of the concomitance of these bacteria and Leishmania parasite outside the midgut environment have not been investigated in the infection process. Herein, a needle infection model was designed to mimic transmission by sand flies, to examine differences in the onset and progression of L. major infection initiated by inoculation with "low" or "high" doses of Enterobacter cloacae and Bacillus subtilis bacteria. The results showed an alteration in the local expression of pro- and anti-inflammatory cytokines in mice receiving different inoculations of bacteria. Simultaneous injection of two bacteria with Leishmania parasites in the low-dose group caused greater thickness of ear pinna and enhanced tissue chronic inflammatory cells, as well as resulted in multifold increase in the expression of IL-4 and IL-1β and a decrease in the iNOS expression, without changing the L. major burden. Despite advances in scientific breakthroughs, scant survey has investigated the interaction between micro and macro levels of organization of leishmaniasis that ranges from the cellular to macro ecosystem levels, giving rise to the spread and persistence of the disease in a region. Our findings provide new insight into using the potential of the vector-derived microbiota in modulating the vertebrate immune system for the benefit of the host or recommend the use of appropriate antibiotics along with antileishmanial medicines.
Collapse
Affiliation(s)
- Fariba Amni
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Naseh Maleki-Ravasan
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| | - Mahmoud Nateghi-Rostami
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| | - Ramtin Hadighi
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| | - Fateh Karimian
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Reza Meamar
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Badirzadeh
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parviz Parvizi
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| |
Collapse
|
43
|
Jeong K, Chang J, Park SM, Kim J, Jeon S, Kim DH, Kim YE, Lee JC, Im S, Jo Y, Min JY, Lee H, Yeom M, Seok SH, On DI, Noh H, Yun JW, Park JW, Song D, Seong JK, Kim KC, Lee JY, Park HJ, Kim S, Nam TG, Lee W. Rapid discovery and classification of inhibitors of coronavirus infection by pseudovirus screen and amplified luminescence proximity homogeneous assay. Antiviral Res 2023; 209:105473. [PMID: 36435212 PMCID: PMC9682871 DOI: 10.1016/j.antiviral.2022.105473] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022]
Abstract
To identify potent antiviral compounds, we introduced a high-throughput screen platform that can rapidly classify hit compounds according to their target. In our platform, we performed a compound screen using a lentivirus-based pseudovirus presenting a spike protein of coronavirus, and we evaluated the hit compounds using an amplified luminescence proximity homogeneous assay (alpha) test with purified host receptor protein and the receptor binding domain of the viral spike. With our screen platform, we were able to identify both spike-specific compounds (class I) and broad-spectrum antiviral compounds (class II). Among the hit compounds, thiosemicarbazide was identified to be selective to the interaction between the viral spike and its host cell receptor, and we further optimized the binding potency of thiosemicarbazide through modification of the pyridine group. Among the class II compounds, we found raloxifene and amiodarone to be highly potent against human coronaviruses including Middle East respiratory syndrome coronavirus (MERS-CoV), severe acute respiratory syndrome coronavirus (SARS-CoV), and SARS-CoV-2. In particular, using analogs of the benzothiophene moiety, which is also present in raloxifene, we have identified benzothiophene as a novel structural scaffold for broad-spectrum antivirals. This work highlights the strong utility of our screen platform using a pseudovirus assay and an alpha test for rapid identification of potential antiviral compounds and their mechanism of action, which can lead to the accelerated development of therapeutics against newly emerging viral infections.
Collapse
Affiliation(s)
- Kwiwan Jeong
- Bio-center, Gyeonggido Business and Science Accelerator, Suwon, South Korea,Corresponding author
| | - JuOae Chang
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sun-mi Park
- Bio-center, Gyeonggido Business and Science Accelerator, Suwon, South Korea
| | - Jinhee Kim
- Institut Pasteur Korea, Seongnam, South Korea
| | | | - Dong Hwan Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Young-Eui Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Joo Chan Lee
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Somyoung Im
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Yejin Jo
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | | | - Hanbyeul Lee
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Minjoo Yeom
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, South Korea
| | - Da In On
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Hyuna Noh
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, South Korea
| | - Daesub Song
- Department of Veterinary Medicine Virology Laboratory, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, South Korea
| | - Kyung-Chang Kim
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Joo-Yeon Lee
- Center for Emerging Virus Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, South Korea
| | - Hyun-Ju Park
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea,Corresponding author
| | - Seungtaek Kim
- Institut Pasteur Korea, Seongnam, South Korea,Corresponding author
| | - Tae-gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea,Corresponding author
| | - Wonsik Lee
- Department of Pharmacy, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea,Corresponding author
| |
Collapse
|
44
|
van Gijsel-Bonnello M, Darling NJ, Tanaka T, Di Carmine S, Marchesi F, Thomson S, Clark K, Kurowska-Stolarska M, McSorley HJ, Cohen P, Arthur JSC. Salt-inducible kinase 2 regulates fibrosis during bleomycin-induced lung injury. J Biol Chem 2022; 298:102644. [PMID: 36309093 PMCID: PMC9706632 DOI: 10.1016/j.jbc.2022.102644] [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: 09/15/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a progressive and normally fatal disease with limited treatment options. The tyrosine kinase inhibitor nintedanib has recently been approved for the treatment of idiopathic pulmonary fibrosis, and its effectiveness has been linked to its ability to inhibit a number of receptor tyrosine kinases including the platelet-derived growth factor, vascular endothelial growth factor, and fibroblast growth factor receptors. We show here that nintedanib also inhibits salt-inducible kinase 2 (SIK2), with a similar IC50 to its reported tyrosine kinase targets. Nintedanib also inhibited the related kinases SIK1 and SIK3, although with 12-fold and 72-fold higher IC50s, respectively. To investigate if the inhibition of SIK2 may contribute to the effectiveness of nintedanib in treating lung fibrosis, mice with kinase-inactive knockin mutations were tested using a model of bleomycin-induced lung fibrosis. We found that loss of SIK2 activity protects against bleomycin-induced fibrosis, as judged by collagen deposition and histological scoring. Loss of both SIK1 and SIK2 activity had a similar effect to loss of SIK2 activity. Total SIK3 knockout mice have a developmental phenotype making them unsuitable for analysis in this model; however, we determined that conditional knockout of SIK3 in the immune system did not affect bleomycin-induced lung fibrosis. Together, these results suggest that SIK2 is a potential drug target for the treatment of lung fibrosis.
Collapse
Affiliation(s)
- Manuel van Gijsel-Bonnello
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom; MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Nicola J Darling
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Takashi Tanaka
- Research Centre of Specialty, Ono Pharmaceutical Co Ltd, Osaka, Japan
| | - Samuele Di Carmine
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Francesco Marchesi
- School of Veterinary Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Thomson
- Biological Services, University of Dundee, Dundee, United Kingdom
| | - Kristopher Clark
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Mariola Kurowska-Stolarska
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
45
|
Şener Akçora D, Erdoğan D, Take Kaplanoğlu G, Göktaş GE, Şeker U, Elmas Ç. Electron microscopic investigation of benzo(a)pyrene-induced alterations in the rat kidney tissue and the protective effects of curcumin. Ultrastruct Pathol 2022; 46:519-530. [DOI: 10.1080/01913123.2022.2152144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dila Şener Akçora
- Department of Histology and Embryology, Faculty of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Deniz Erdoğan
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Turkey
| | | | - Gül Eser Göktaş
- Department of Histology and Embryology, Faculty of Medicine, Lokman Hekim University, Turkey
| | - Uğur Şeker
- Department of Histology and Embryology, Faculty of Medicine, Harran University, Sanlıurfa, Turkey
| | - Çiğdem Elmas
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Turkey
| |
Collapse
|
46
|
Lin CC, Chuang KC, Chen SW, Chao YH, Yen CC, Yang SH, Chen W, Chang KH, Chang YK, Chen CM. Lactoferrin Ameliorates Ovalbumin-Induced Asthma in Mice through Reducing Dendritic-Cell-Derived Th2 Cell Responses. Int J Mol Sci 2022; 23:ijms232214185. [PMID: 36430662 PMCID: PMC9696322 DOI: 10.3390/ijms232214185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/18/2022] Open
Abstract
Asthma is a chronic respiratory disease with symptoms such as expiratory airflow narrowing and airway hyperresponsiveness (AHR). Millions of people suffer from asthma and are at risk of life-threatening conditions. Lactoferrin (LF) is a glycoprotein with multiple physiological functions, including antioxidant, anti-inflammatory, antimicrobial, and antitumoral activities. LF has been shown to function in immunoregulatory activities in ovalbumin (OVA)-induced delayed type hypersensitivity (DTH) in mice. Hence, the purpose of this study was to investigate the roles of LF in AHR and the functions of dendritic cells (DCs) and Th2-related responses in asthma. Twenty 8-week-old male BALB/c mice were divided into normal control (NC), ovalbumin (OVA)-sensitized, and OVA-sensitized with low dose of LF (100 mg/kg) or high dose of LF (300 mg/kg) treatment groups. The mice were challenged by intranasal instillation with 5% OVA on the 21st to 27th day after the start of the sensitization period. The AHR, cytokines in bronchoalveolar lavage fluid, and pulmonary histology of each mouse were measured. Serum OVA-specific IgE and IgG1 and OVA-specific splenocyte responses were further detected. The results showed that LF exhibited protective effects in ameliorating AHR, as well as lung inflammation and damage, in reducing the expression of Th2 cytokines and the secretion of allergen-specific antibodies, in influencing the functions of DCs, and in decreasing the level of Th2 immune responses in a BALB/c mouse model of OVA-induced allergic asthma. Importantly, we demonstrated that LF has practical application in reducing DC-induced Th2 cell responses in asthma. In conclusion, LF exhibits anti-inflammation and immunoregulation activities in OVA-induced allergic asthma. These results suggest that LF may act as a supplement to prevent asthma-induced lung injury and provide an additional agent for reducing asthma severity.
Collapse
Affiliation(s)
- Chi-Chien Lin
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kai-Cheng Chuang
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Shih-Wei Chen
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Otolaryngology, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Ya-Hsuan Chao
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Chih-Ching Yen
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Internal Medicine, China Medical University Hospital, College of Health Care, China Medical University, Taichung 404, Taiwan
| | - Shang-Hsun Yang
- Department of Physiology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Wei Chen
- Division of Pulmonary and Critical Care Medicine, Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Kuang-Hsi Chang
- Department of Medical Research, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Yu-Kang Chang
- Department of Medical Research, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- The iEGG and Animal Biotechnology Center, The Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Correspondence: ; Tel.: +886-4-22856309; Fax: +886-4-22874740
| |
Collapse
|
47
|
Le‐Trilling VTK, Ebel J, Baier F, Wohlgemuth K, Pfeifer KR, Mookhoek A, Krebs P, Determann M, Katschinski B, Adamczyk A, Lange E, Klopfleisch R, Lange CM, Sokolova V, Trilling M, Westendorf AM. Acute cytomegalovirus infection modulates the intestinal microbiota and targets intestinal epithelial cells. Eur J Immunol 2022. [DOI: 10.1002/eji.202249940 10.1002/eji.202249940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
| | - Jana‐Fabienne Ebel
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Franziska Baier
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Kerstin Wohlgemuth
- Institute for Virology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Kai Robin Pfeifer
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Aart Mookhoek
- Institute of Pathology University of Bern Bern Switzerland
| | - Philippe Krebs
- Institute of Pathology University of Bern Bern Switzerland
| | - Madita Determann
- Institute for Virology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Benjamin Katschinski
- Institute for Virology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Alexandra Adamczyk
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Erik Lange
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology Free University of Berlin Berlin Germany
| | - Christian M. Lange
- Department of Gastroenterology and Hepatology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Viktoriya Sokolova
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
- Inorganic Chemistry and Centre for Nanointegration Duisburg‐Essen (CeNIDE) University of Duisburg‐Essen Essen Germany
| | - Mirko Trilling
- Institute for Virology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology University Hospital Essen University of Duisburg‐Essen Essen Germany
| |
Collapse
|
48
|
Luo T, Wang D, Zhao Y, Li X, Yang G, Jin Y. Polystyrene microplastics exacerbate experimental colitis in mice tightly associated with the occurrence of hepatic inflammation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 844:156884. [PMID: 35752249 DOI: 10.1016/j.scitotenv.2022.156884] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 05/27/2023]
Abstract
The potential health effects of microplastics (MPs) have become a public concern due to their ubiquitousness in the environment and life. Numerous studies have demonstrated that a high dose of MPs can adversely affect gastrointestinal health. However, few studies have focused on the impact of microplastics on patients' health with respect to gastrointestinal diseases. Inflammatory bowel disease (IBD) has emerged as a global disease with a rapidly increasing incidence. IBD, a specific gastrointestinal illness characterized by acute, chronic inflammation and intestinal barrier dysfunction, might increase sensitivity to MPs exposure. Herein, we investigated the impact and mechanism of PS-MPs on dextran sodium sulfate (DSS)-induced colitis. The results demonstrated that gavage with PS-MPs alone caused minimal effects on the intestinal barrier and liver status of mice. For mice with colitis, additional PS-MPs exposure caused a shorter colon length, aggravated histopathological damage and inflammation, reduced mucus secretion, and increased the colon permeability. Furthermore, PS-MPs exposure also increased the risk of secondary liver injury associated with inflammatory cell infiltration. These findings provide more histopathological evidence and suggest a need for more research on the health risk of MPs for sensitive individuals.
Collapse
Affiliation(s)
- Ting Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Yao Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Xinfang Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Guiling Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China.
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
49
|
A single-administration therapeutic interfering particle reduces SARS-CoV-2 viral shedding and pathogenesis in hamsters. Proc Natl Acad Sci U S A 2022; 119:e2204624119. [PMID: 36074824 PMCID: PMC9522362 DOI: 10.1073/pnas.2204624119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The high transmissibility of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a primary driver of the COVID-19 pandemic. While existing interventions prevent severe disease, they exhibit mixed efficacy in preventing transmission, presumably due to their limited antiviral effects in the respiratory mucosa, whereas interventions targeting the sites of viral replication might more effectively limit respiratory virus transmission. Recently, intranasally administered RNA-based therapeutic interfering particles (TIPs) were reported to suppress SARS-CoV-2 replication, exhibit a high barrier to resistance, and prevent serious disease in hamsters. Since TIPs intrinsically target the tissues with the highest viral replication burden (i.e., respiratory tissues for SARS-CoV-2), we tested the potential of TIP intervention to reduce SARS-CoV-2 shedding. Here, we report that a single, postexposure TIP dose lowers SARS-CoV-2 nasal shedding, and at 5 days postinfection, infectious virus shed is below detection limits in 4 out of 5 infected animals. Furthermore, TIPs reduce shedding of Delta variant or WA-1 from infected to uninfected hamsters. Cohoused "contact" animals exposed to infected, TIP-treated animals exhibited significantly lower viral loads, reduced inflammatory cytokines, no severe lung pathology, and shortened shedding duration compared to animals cohoused with untreated infected animals. TIPs may represent an effective countermeasure to limit SARS-CoV-2 transmission.
Collapse
|
50
|
A single-administration therapeutic interfering particle reduces SARS-CoV-2 viral shedding and pathogenesis in hamsters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022. [PMID: 35982679 DOI: 10.1101/2022.08.10.503534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The high transmissibility of SARS-CoV-2 is a primary driver of the COVID-19 pandemic. While existing interventions prevent severe disease, they exhibit mixed efficacy in preventing transmission, presumably due to their limited antiviral effects in the respiratory mucosa, whereas interventions targeting the sites of viral replication might more effectively limit respiratory virus transmission. Recently, intranasally administered RNA-based therapeutic interfering particles (TIPs) were reported to suppress SARS-CoV-2 replication, exhibit a high barrier to resistance, and prevent serious disease in hamsters. Since TIPs intrinsically target the tissues with the highest viral replication burden (i.e., respiratory tissues for SARS-CoV-2), we tested the potential of TIP intervention to reduce SARS-CoV-2 shedding. Here, we report that a single, post-exposure TIP dose lowers SARS-CoV-2 nasal shedding and at 5 days post-infection infectious virus shed is below detection limits in 4 out of 5 infected animals. Furthermore, TIPs reduce shedding of Delta variant or WA-1 from infected to uninfected hamsters. Co-housed 'contact' animals exposed to infected, TIP-treated, animals exhibited significantly lower viral loads, reduced inflammatory cytokines, no severe lung pathology, and shortened shedding duration compared to animals co-housed with untreated infected animals. TIPs may represent an effective countermeasure to limit SARS-CoV-2 transmission. Significance COVID-19 vaccines are exceptionally effective in preventing severe disease and death, but they have mixed efficacy in preventing virus transmission, consistent with established literature that parenteral vaccines for other viruses fail to prevent mucosal virus shedding or transmission. Likewise, small-molecule antivirals, while effective in reducing viral-disease pathogenesis, also appear to have inconsistent efficacy in preventing respiratory virus transmission including for SARS-CoV-2. Recently, we reported the discovery of a single-administration antiviral Therapeutic Interfering Particle (TIP) against SARS-CoV-2 that prevents severe disease in hamsters and exhibits a high genetic barrier to the evolution of resistance. Here, we report that TIP intervention also reduces SARS-CoV-2 transmission between hamsters.
Collapse
|