1
|
Baradaran B, Hazrati A, Kazemi-Sefat NA, Soleimanjahi H, Soudi S. Umbilical cord-derived mesenchymal stem cell condition medium effect on rotavirus-infected Caco-2 cells survival and inflammatory responses. Tissue Cell 2025; 93:102699. [PMID: 39818065 DOI: 10.1016/j.tice.2024.102699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
Rotavirus is the most important cause of severe gastroenteritis in infants and children worldwide. This virus causes an increase in inflammatory responses by increasing cellular oxidative stress and the expression and activity of the transcription factor NF-κB and COX-2. As a result of NF-κB activation, the expression of inflammatory cytokines also increases. So, there is a need to control pathogenic inflammatory responses mediated by rotavirus. Mesenchymal stem cells (MSCs) have confirmed immunomodulatory characteristics. The present study aims to investigate the effects of MSCs conditioned media (MSCs-CM) in reducing the inflammatory response of Caco-2 cells when exposed to rotavirus. 72 h After rotavirus-infected Caco-2 cell of treatment with MSCs-CM, virus replication (CCID50), secretion of IL-6, and IL-8 (ELISA), COX-2 and NF-κB genes expression (q-PCR), apoptosis (Annexin V-PI), and nitric oxide (NO) level (Gries's reagent) are investigated. Based on the results, virus replication was reduced by Log1 in the CM-treated groups. Also, treating Caco-2 cells with MSCs-CM led to decreased IL-6 and NO and increased IL-8 production. Evaluation of apoptosis in MSCs-CM-treated rotavirus-exposed Caco-2 cells showed a significant reduction in their apoptosis. Also, the expression of COX-2 is increased significantly. However, the expression of NF-κB decreased significantly after treatment with MSCs-CM. The results show that inflammatory responses, oxidative stress, and apoptosis in rotavirus-infected cells have decreased after treatment with MSC-CM.
Collapse
Affiliation(s)
- Behnoosh Baradaran
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Nguyen HT, Hoa-Tran TN, Tran HQ, Nguyen TTT. In Vitro Inhibitory Effect of Berberine Against Rotavirus. Chem Biodivers 2025; 22:e202400986. [PMID: 39400499 DOI: 10.1002/cbdv.202400986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Although berberine (BBR) is well known as an active constituent in traditional medicines used in the treatment of gastrointestinal diseases, its potential against viral gastroenteritis has not been specifically reported. This study aims to investigate the antiviral activity of BBR against rotavirus and evaluate its cytotoxicity and pharmacological efficacies, including antioxidant and anti-inflammatory activities in vitro. Using ultraviolet-visible absorption spectroscopy, the saturation concentration of BBR was determined as 2261 μg/mL, indicating that BBR is a poor water-soluble compound. The inhibition rate of nitric oxide (NO) production of BBR solution at a concentration of 283 μg/mL was similar to that of Cardamonin 0.3 μM with a cell viability of 92.46±0.35 %, revealing the anti-inflammatory activity of BBR. The cytotoxicity of the BBR solution depended on its concentration, whereby the 50 % cytotoxicity concentration (CC50) of BBR after 96 h exposure was 664 μg/mL. Investigation of cytopathic effects (CPEs) of MA104 cells treated with BBR and BBR-incubated rotavirus indicates that BBR could effectively inhibit the replication of rotavirus. CPEs were not observed in the cells inoculated with rotavirus (100TCID50) which was pre-incubated with BBR for 96 hours at a BBR concentration of 283 μg/mL. Therefore, the study provides reliable results to demonstrate the ability of BBR to inhibit the replication of rotavirus.
Collapse
Affiliation(s)
- Hue Thi Nguyen
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, 12116, Vietnam
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, 12116, Vietnam
| | - Thi Nguyen Hoa-Tran
- Department of Virology, National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Huy Quang Tran
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, 12116, Vietnam
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, 12116, Vietnam
| | - Thuy Thi Thu Nguyen
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, 12116, Vietnam
| |
Collapse
|
3
|
Zhao R, Jiang C, Yuan Y, Zhang S, Ghonaim AH, Che C, Li X, Jin M, Jin E, Zeng X, Li S, Ren M. Isoleucine Enhanced the Function of the Small Intestinal Mucosal Barrier in Weaned Piglets to Alleviate Rotavirus Infection. Animals (Basel) 2024; 14:3146. [PMID: 39518871 PMCID: PMC11545378 DOI: 10.3390/ani14213146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Rotavirus (RV) is a major cause of diarrhea in young children and animals, especially piglets, leading to substantial economic losses in the global pig industry. Isoleucine (Ile), a branched-chain amino acid, plays an important role in regulating nutrient metabolism and has been shown to improve diarrhea. This study aimed to evaluate the effects of Ile supplementation on the mucosal immune barrier of the small intestine in RV-infected weaned piglets. METHODS Forty-eight 21-day-old weaned piglets were randomly divided into three dietary treatments (each treatment was subdivided into two groups, eight replicates per group), with 0%, 0.5%, or 1% Ile added for 15 days, and then, one group from each treatment was challenged with RV. RESULTS The results showed that 1% Ile added to the diet promoted the healthy development of the intestinal mucosa. Ile could restore the reduced villus height in the ileum and the goblet cell number in the duodenum and ileum to normal levels, improving the intestinal epithelial tight junctions in RV-infected piglets. Additionally, Ile increased the activity of lipase, amylase, and sucrase, as well as superoxide dismutase (SOD) and glutathione (GSH), along with the expression of SIgA, DEFβ1, and DEFβ2 in parts of the small intestine. CONCLUSIONS The addition of Ile to the diet mitigated the effects of RV infection on intestinal morphology and mucosal barrier function, as well as the physiological functions of weaned piglets, and improved the antioxidant and immune functions of the piglets to some extent. These findings offer valuable insights, contributing to a deeper understanding of the role of Ile in supporting intestinal health.
Collapse
Affiliation(s)
- Rongkun Zhao
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Changsheng Jiang
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Yuchen Yuan
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Shen Zhang
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Ahmed H. Ghonaim
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Desert Research Center, Cairo 11435, Egypt
| | - Chuanyan Che
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Xiaojin Li
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Mengmeng Jin
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Erhui Jin
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Shenghe Li
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Man Ren
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| |
Collapse
|
4
|
Mukherjee A, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial Reactive Oxygen Species in Infection and Immunity. Biomolecules 2024; 14:670. [PMID: 38927073 PMCID: PMC11202257 DOI: 10.3390/biom14060670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Reactive oxygen species (ROS) contain at least one oxygen atom and one or more unpaired electrons and include singlet oxygen, superoxide anion radical, hydroxyl radical, hydroperoxyl radical, and free nitrogen radicals. Intracellular ROS can be formed as a consequence of several factors, including ultra-violet (UV) radiation, electron leakage during aerobic respiration, inflammatory responses mediated by macrophages, and other external stimuli or stress. The enhanced production of ROS is termed oxidative stress and this leads to cellular damage, such as protein carbonylation, lipid peroxidation, deoxyribonucleic acid (DNA) damage, and base modifications. This damage may manifest in various pathological states, including ageing, cancer, neurological diseases, and metabolic disorders like diabetes. On the other hand, the optimum levels of ROS have been implicated in the regulation of many important physiological processes. For example, the ROS generated in the mitochondria (mitochondrial ROS or mt-ROS), as a byproduct of the electron transport chain (ETC), participate in a plethora of physiological functions, which include ageing, cell growth, cell proliferation, and immune response and regulation. In this current review, we will focus on the mechanisms by which mt-ROS regulate different pathways of host immune responses in the context of infection by bacteria, protozoan parasites, viruses, and fungi. We will also discuss how these pathogens, in turn, modulate mt-ROS to evade host immunity. We will conclude by briefly giving an overview of the potential therapeutic approaches involving mt-ROS in infectious diseases.
Collapse
Affiliation(s)
- Arunima Mukherjee
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| |
Collapse
|
5
|
Cao J, Wen Z, Zhang Y, Zhang B, Chen Y, Xing G, Wu Y, Zhou Z, Liu X, Hou S. Effects of DHAV-3 infection on innate immunity, antioxidant capacity, and lipid metabolism in ducks with different DHAV-3 susceptibilities. Poult Sci 2024; 103:103374. [PMID: 38295495 PMCID: PMC10844866 DOI: 10.1016/j.psj.2023.103374] [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: 09/19/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 02/02/2024] Open
Abstract
The aim of the experiment was to evaluate the status of innate immunity, oxidative status and lipid accumulation in ducklings exhibiting varying susceptibilities to DHAV-3 infection. In the experiment, ducklings with different DHAV-3 susceptibilities were used. Samples were collected at 6, 12, 15, and 24 h post infection (hpi), with 5 samples per time point. Plasma biochemistry, antioxidant enzyme activities, lipid content of liver and kidney were detected in the experiment. Elevated plasma level of total bilirubin, direct bilirubin, and creatinine indicated the injury of liver and kidney in susceptible ducklings (P < 0.05). The histopathological sections showed the injury in kidney. During the infection time, there was an increase in the concentrations of reactive oxygen species and oxidative damage markers (malondialdehyde and nitric oxide) in plasma of susceptible ducklings, particularly at 24 hpi (P < 0.05). Compared with the resistant ducklings, DHAV-3 infection resulted in a significant increase in the plasma total triglyceride (TG) level and a decrease in glucose level in susceptible ducklings. Gene expression of the innate immune response was both investigated in liver and kidney. In resistant ducklings, the expressions levels of pattern recognition receptors RIG-I, MDA5 remained constant. In contrast, the gene expressions peaked at 24 hpi in the susceptible ducklings. DHAV-3 infection promoted the expression of IFN, IL6, IL12β, caspase-8 or caspase-9 in both liver and kidney of susceptible ducklings. In conclusion, DHAV-3 infection led to the mobilization of antioxidant defenses, alterations in lipid metabolism, and oxidative stress in susceptible ducklings during DHAV-3 infection.
Collapse
Affiliation(s)
- Junting Cao
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhiguo Wen
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yunsheng Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bo Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ying Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guangnan Xing
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yongbao Wu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhengkui Zhou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaolin Liu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Shuisheng Hou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
6
|
Lai X, Wu A, Yu B, Yan H, Luo J, Zheng P, Yu J, Chen D. Retinoic acid alleviates rotavirus-induced intestinal damage by regulating redox homeostasis and autophagic flux in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:409-421. [PMID: 38371474 PMCID: PMC10874719 DOI: 10.1016/j.aninu.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 02/20/2024]
Abstract
Rotaviruses (RV) are a major cause of severe gastroenteritis, particularly in neonatal piglets. Despite the availability of effective vaccines, the development of antiviral therapies for RV remains an ongoing challenge. Retinoic acid (RA), a metabolite of vitamin A, has been shown to have anti-oxidative and antiviral properties. However, the mechanism by which RA exerts its intestinal-protective and antiviral effects on RV infection is not fully understood. The study investigates the effects of RA supplementation in Duroc × Landrace × Yorkshire (DLY) piglets challenged with RV. Thirty-six DLY piglets were assigned into six treatments, including a control group, RA treatment group with two concentration gradients (5 and 15 mg/d), RV treatment group, and RV treatment group with the addition of different concentration gradients of RA (5 and 15 mg/d). Our study revealed that RV infection led to extensive intestinal architecture damage, which was mitigated by RA treatment at lower concentrations by increasing the villus height and villus height/crypt depth ratio (P < 0.05), enhancing intestinal stem cell signaling and promoting intestinal barrier functions. In addition, 15 mg/d RA supplementation significantly increased NRF2 and HO-1 protein expression (P < 0.05) and GSH content (P < 0.05), indicating that RA supplementation can enhance anti-oxidative signaling and redox homeostasis after RV challenge. Additionally, the research demonstrated that RA exerts a dual impact on the regulation of autophagy, both stimulating the initiation of autophagy and hindering the flow of autophagic flux. Through the modulation of autophagic flux, RA influence the progression of RV infection. These findings provide new insights into the regulation of redox hemostasis and autophagy by RA and its potential therapeutic application in RV infection.
Collapse
Affiliation(s)
- Xin Lai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
- Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
7
|
Guerrero M, Hernández J, Gomez L, Guerrero C. Oxidative stress enhances rotavirus oncolysis in breast cancer and leukemia, except in melanoma with abundant matrix. Virus Res 2024; 339:199285. [PMID: 38013142 PMCID: PMC10711233 DOI: 10.1016/j.virusres.2023.199285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVES This study aimed to explore the impact of oxidative stress and extracellular matrix integrity on rotavirus infection in various cancer cells, including breast cancer, acute lymphoblastic leukemia, and melanoma. METHODS We induced oxidative stress using ROS-inducing drugs (cisplatin, metronidazole, melatonin, valproic acid, doxorubicin, losartan, nitrofurantoin, and DHA) and investigated the effects on viral infection in MCF-7, Reh, A375, B16-F1, and SK-MEL-28 cells and the generation of virions from infected cells by harvesting the supernatants every two hours, reinfecting other cells, and analyzing cell viability and DNA fragmentation. FINDINGS In MCF-7 and Reh cells, rotavirus Wt1-5 infection led to increased ROS generation, virion production, membrane permeability, mitochondrial dysfunction, DNA damage, and cell death. These effects were amplified by ROS-inducing drugs. Conversely, melanoma cells (SK-MEL-28 and A375) with a robust extracellular matrix network showed limited sensitivity to the drugs. Notably, losartan, which modulates the extracellular matrix, enhanced viral infection in melanoma cells (99 %). CONCLUSIONS Oxidative stress promotes oncolytic rotavirus infection in breast cancer and acute lymphoblastic leukemia cells, suggesting potential utility in combination with radiotherapy or chemotherapy due to their shared induction of intracellular oxidative stress.
Collapse
Affiliation(s)
- Marvi Guerrero
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad Nacional de Colombia, DC, Bogotá, Colombi
| | - Juan Hernández
- Grupo de Fisiología Molecular del Instituto Nacional de Salud. A. A. 80080. Av. Calle 26 No. 51-20 DC, Bogotá, Colombia
| | - Luis Gomez
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad Nacional de Colombia, DC, Bogotá, Colombi; Grupo de Fisiología Molecular del Instituto Nacional de Salud. A. A. 80080. Av. Calle 26 No. 51-20 DC, Bogotá, Colombia
| | - Carlos Guerrero
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad Nacional de Colombia, DC, Bogotá, Colombi.
| |
Collapse
|
8
|
Banerjee S, Sarkar R, Mukherjee A, Mitra S, Gope A, Chawla-Sarkar M. Rotavirus-induced lncRNA SLC7A11-AS1 promotes ferroptosis by targeting cystine/glutamate antiporter xCT (SLC7A11) to facilitate virus infection. Virus Res 2024; 339:199261. [PMID: 37923170 PMCID: PMC10684390 DOI: 10.1016/j.virusres.2023.199261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
Rotavirus (RV) is the primary etiological agent of virus-associated gastroenteritis in infants, causing 200,000 childhood death annually. Despite the availability of vaccines, rotaviral diarrhea continues to be a severe issue in underdeveloped nations in Asia and Africa. The situation demands continual studies on host-rotavirus interactions to understand disease pathogenesis and develop effective antiviral therapeutics. Long non-coding RNAs (lncRNAs), which are a subset of non-coding RNAs of more than 200 nucleotides in length, are reported to play a regulatory function in numerous viral infections. Virus infection often alters the host transcriptome including lncRNA that are differentially expressed either to play an antiviral role or to be advantageous towards virus propagation. In the current study, qPCR array-based expression profiling of host lncRNAs was performed in rotavirus-infected HT-29 cells that identified the lncRNA SLC7A11-AS1 to be upregulated during RV infection. Knockdown of SLC7A11-AS1 conspicuously reduced RV titers implying its pro-viral significance. RV-induced SLC7A11-AS1 downregulates the gene SLC7A11/xCT that encodes the light chain subunit of the system XC- cystine-glutamate exchange transporter, leading to decrease in intracellular glutathione level and increase in lipid peroxidation, which are signature features of ferroptotic pathway. Ectopic expression of xCT also abrogated RV infection by reversing the virus optimized levels of intracellular GSH and lipid ROS levels. Cumulatively, the study reveals that RV infection triggers ferroptotic cell death via SLC7A11-AS1/xCT axis to facilitate its own propagation.
Collapse
Affiliation(s)
- Shreya Banerjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Rakesh Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Arpita Mukherjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Suvrotoa Mitra
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Animesh Gope
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, P-33, C.I.T. Road, Scheme-XM, Beliaghata, Kolkata, West Bengal 700010, India.
| |
Collapse
|
9
|
Pincemail J, Rousseau AF, Kaux JF, Cheramy-Bien JP, Bruyère C, Prick J, Stern D, Kaci MM, Maertens De Noordhout B, Albert A, Eubelen C, Goff CL, Misset B, Cavalier E, Charlier C, Meziane S. A Pilot Study on Oxidative Stress during the Recovery Phase in Critical COVID-19 Patients in a Rehabilitation Facility: Potential Utility of the PAOT ® Technology for Assessing Total Anti-Oxidative Capacity. Biomedicines 2023; 11:biomedicines11051308. [PMID: 37238982 DOI: 10.3390/biomedicines11051308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Oxidative stress (OS) could cause various COVID-19 complications. Recently, we have developed the Pouvoir AntiOxydant Total (PAOT®) technology for reflecting the total antioxidant capacity (TAC) of biological samples. We aimed to investigate systemic oxidative stress status (OSS) and to evaluate the utility of PAOT® for assessing TAC during the recovery phase in critical COVID-19 patients in a rehabilitation facility. MATERIALS AND METHODS In a total of 12 critical COVID-19 patients in rehabilitation, 19 plasma OSS biomarkers were measured: antioxidants, TAC, trace elements, oxidative damage to lipids, and inflammatory biomarkers. TAC level was measured in plasma, saliva, skin, and urine, using PAOT and expressed as PAOT-Plasma, -Saliva, -Skin, and -Urine scores, respectively. Plasma OSS biomarker levels were compared with levels from previous studies on hospitalized COVID-19 patients and with the reference population. Correlations between four PAOT scores and plasma OSS biomarker levels were analyzed. RESULTS During the recovery phase, plasma levels in antioxidants (γ-tocopherol, β-carotene, total glutathione, vitamin C and thiol proteins) were significantly lower than reference intervals, whereas total hydroperoxides and myeloperoxidase (a marker of inflammation) were significantly higher. Copper negatively correlated with total hydroperoxides (r = 0.95, p = 0.001). A similar, deeply modified OSS was already observed in COVID-19 patients hospitalized in an intensive care unit. TAC evaluated in saliva, urine, and skin correlated negatively with copper and with plasma total hydroperoxides. To conclude, the systemic OSS, determined using a large number of biomarkers, was always significantly increased in cured COVID-19 patients during their recovery phase. The less costly evaluation of TAC using an electrochemical method could potentially represent a good alternative to the individual analysis of biomarkers linked to pro-oxidants.
Collapse
Affiliation(s)
- Joël Pincemail
- Clinical Chemistry, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | | | - Jean-François Kaux
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Jean-Paul Cheramy-Bien
- Department of Cardiovascular Surgery, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Christine Bruyère
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Jeanine Prick
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - David Stern
- Veterinary Medicine Faculty, FARAH, University of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Mouna-Messaouda Kaci
- Research Department, Institut Européen des Antioxydants (IEA), Oxystress Technologies PAOTScan, 54500 Vandœuvre-lès-Nancy, France
| | - Benoît Maertens De Noordhout
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Adelin Albert
- Biostatistics Department, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Céline Eubelen
- Physical Medicine Rehabilitation and Sports Traumatology Department Sports, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Caroline Le Goff
- Clinical Chemistry, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Benoît Misset
- Intensive Care Department, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Etienne Cavalier
- Clinical Chemistry, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Corinne Charlier
- Toxicology Department, University Hospital of Liège, Sart Tilman, 4000 Liège, Belgium
| | - Smail Meziane
- Research Department, Institut Européen des Antioxydants (IEA), Oxystress Technologies PAOTScan, 54500 Vandœuvre-lès-Nancy, France
| |
Collapse
|
10
|
Lee GA, Chang YW, Lin WL, Yang YCSH, Chen WJ, Huang FH, Liu YR. Modulatory Effects of Heat-Inactivated Streptococcus Thermophilus Strain 7 on the Inflammatory Response: A Study on an Animal Model with TLR3-Induced Intestinal Injury. Microorganisms 2023; 11:microorganisms11020278. [PMID: 36838243 PMCID: PMC9959611 DOI: 10.3390/microorganisms11020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Rotavirus infections result in severe gastroenteritis with a detrimental inflammatory response in the intestine. Because probiotics have an anti-inflammatory effect and can modulate the gut microbiota profile, they can be used as a biotherapy for inflammatory intestinal diseases. In this study, we isolated Streptococcus thermophilus strain 7 (ST7) from cow milk and examined the effect of heat-inactivated ST7 on the intestinal histopathological score, inflammatory cytokine levels, T-cell activation and effector function, and microbiome profile in a mouse model with intestinal injury induced by polyinosinic-polycytidylic acid (poly I:C), a Toll-like receptor 3 agonist. The results indicated that ST7 treatment prevented weight loss and intestinal injury and prevented the upregulation of serum interleukin-6 (IL-6), tumor necrosis factor-α, and IL-15 levels in intestinal epithelial cells; prevented the upregulation of inflammation-associated Gammaproteobacteria and Alistipes; and increased the levels of Firmicutes in fecal microbiota after poly I:C stimulation. ST7 treatment also increased the serum interferon-γ (IFN-γ) level and promoted the expression of IFN-γ in both CD8 and CD4 T cells. In summary, ST7 prevented the inflammatory response, promoted the T-cell effector function, and modulated the microbiota profile of mice with poly I:C-induced small intestine injury.
Collapse
Affiliation(s)
- Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei City 110, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Child Development Research Center, Taipei Medical University Hospital, Taipei City 110, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Correspondence:
| | - Yu-Wei Chang
- Department of Medical Research, Taipei Medical University Hospital, Taipei City 110, Taiwan
| | - Wan-Li Lin
- Department of Medical Research, Taipei Medical University Hospital, Taipei City 110, Taiwan
| | - Yu-Chen S. H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei City 110, Taiwan
| | - Wei-Jen Chen
- Syngen Biotech International, Shah Alam 40460, Malaysia
| | - Fu-Huan Huang
- Division of Pediatric Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei City 110, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei City 110, Taiwan
| |
Collapse
|
11
|
Behzadi A, imani S, Deravi N, Mohammad Taheri Z, mohammadian F, moraveji Z, Shavysi S, Mostafaloo M, Soleimani Hadidi F, Nanbakhsh S, Olangian-Tehrani S, Marabi MH, behshood P, Poudineh M, Kheirandish A, Keylani K, Behfarnia P. Antiviral Potential of Melissa officinalis L.: A Literature Review. Nutr Metab Insights 2023; 16:11786388221146683. [PMID: 36655201 PMCID: PMC9841880 DOI: 10.1177/11786388221146683] [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: 04/13/2022] [Accepted: 12/05/2022] [Indexed: 01/13/2023] Open
Abstract
The use of synthetic drugs has increased in recent years; however, herbal medicine is yet more trusted among a huge population worldwide; This could be due to minimal side effects, affordable prices, and traditional beliefs. Lemongrass (Melissa officinalis) has been widely used for reducing stress and anxiety, increasing appetite and sleep, reducing pain, healing wounds, and treating poisonous insect bites and bee stings for a long time. Today, research has shown that this plant can also fight viruses including Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), Herpes Simplex Virus (HSV), and Human Immunodeficiency Virus (HIV) through various mechanisms such as inhibiting HSV-1 from binding to host cell, inhibiting HSV-1 replication during the post-adsorption or inhibiting main protease and spike protein of SARS-CoV-2, furthermore, be effective in treating related diseases. This Review investigated the antiviral properties of Melissa officinalis and its effect on viral diseases. More in vitro and in vivo studies are needed to determine Melissa officinaliss underlying mechanism, and more randomized controlled trials should be done to identify its effect in humans. Also, due to the usefulness and lack of side effects, it can be used more as a complementary medicine.
Collapse
Affiliation(s)
- Amirhossein Behzadi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Avicennet, Tehran, Iran
| | - Sadegh imani
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - fatemeh mohammadian
- Student Research Committee, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - zahra moraveji
- Student Research Committee, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sepideh Shavysi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Motahareh Mostafaloo
- School of Nursing and Midwifery, Iran University of Medical Science, Tehran, Iran
| | - Fateme Soleimani Hadidi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Nanbakhsh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Avicennet, Tehran, Iran
| | - Sepehr Olangian-Tehrani
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Avicennet, Tehran, Iran
| | - Mohammad Hesam Marabi
- Student Research Committee, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa behshood
- Department of Microbiology, Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Isfahan, Iran
| | | | - Ali Kheirandish
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kimia Keylani
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pooya Behfarnia
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Ramírez MC, Méndez K, Castelblanco-Mora A, Quijano S, Ulloa J. In Vitro Evaluation of Anti-Rotaviral Activity and Intestinal Toxicity of a Phytotherapeutic Prototype of Achyrocline bogotensis (Kunth) DC. Viruses 2022; 14:v14112394. [PMID: 36366492 PMCID: PMC9695875 DOI: 10.3390/v14112394] [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: 08/17/2022] [Revised: 09/18/2022] [Accepted: 09/29/2022] [Indexed: 01/31/2023] Open
Abstract
Viruses represent the primary etiologic agents (70-80%) of acute diarrheal disease (ADD), and rotavirus (RV) is the most relevant one. Currently, four rotavirus vaccines are available. However, these vaccines do not protect against emerging viral strains or are not available in low-income countries. To date, there are no approved drugs available against rotavirus infection. In this study, we evaluated the in vitro anti-rotaviral activity and intestinal toxicity of a phytotherapeutic prototype obtained from Achyrocline bogotensis (Kunth) DC. (PPAb); medicinal plant that contains compounds that inhibit the rotavirus replication cycle. Virucidal and viral yield reduction effects exerted by the PPAb were evaluated by immunocytochemistry and flow cytometry. Furthermore, the toxic impact of the PPAb was evaluated in polarized human intestinal epithelial C2BBe1 cells in terms of cytotoxicity, loss of cytoplasmic membrane asymmetry, and DNA fragmentation by MTT and fluorometry. PPAb concentrations under 0.49 mg/mL exerted significant virucidal and viral yield reduction activities, and concentrations under 16 mg/mL neither reduced cell viability, produced DNA fragmentation, nor compromised the C2BBe1cell membrane stability after 24-h incubation. Based on these results, the evaluated phytotherapeutic prototype of Achyrocline bogotensis might be considered as a promising alternative to treat ADD caused by rotavirus.
Collapse
Affiliation(s)
- María-Camila Ramírez
- Laboratorio de Virología, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 43-82, Bogotá D.C. 110231, Colombia
| | - Kelly Méndez
- Laboratorio de Virología, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 43-82, Bogotá D.C. 110231, Colombia
| | - Alicia Castelblanco-Mora
- Laboratorio de Virología, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 43-82, Bogotá D.C. 110231, Colombia
| | - Sandra Quijano
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 43-82, Bogotá D.C. 110231, Colombia
| | - Juan Ulloa
- Laboratorio de Virología, Grupo de Enfermedades Infecciosas, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Cra. 7 # 43-82, Bogotá D.C. 110231, Colombia
- Correspondence: ; Tel.: +57-601-3208320 (ext. 4029)
| |
Collapse
|
13
|
Bruno C, Paparo L, Pisapia L, Romano A, Cortese M, Punzo E, Berni Canani R. Protective effects of the postbiotic deriving from cow's milk fermentation with L. paracasei CBA L74 against Rotavirus infection in human enterocytes. Sci Rep 2022; 12:6268. [PMID: 35428750 PMCID: PMC9012738 DOI: 10.1038/s41598-022-10083-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/11/2022] [Indexed: 01/17/2023] Open
Abstract
Rotavirus (RV) is the leading cause of acute gastroenteritis-associated mortality in early childhood. Emerging clinical evidence suggest the efficacy of the postbiotic approach based on cow's milk fermentation with the probiotic Lacticaseibacillus paracasei CBAL74 (FM-CBAL74) in preventing pediatric acute gastroenteritis, but the mechanisms of action are still poorly characterized. We evaluated the protective action of FM-CBAL74 in an in vitro model of RV infection in human enterocytes. The number of infected cells together with the relevant aspects of RV infection were assessed: epithelial barrier damage (tight-junction proteins and transepithelial electrical resistance evaluation), and inflammation (reactive oxygen species, pro-inflammatory cytokines IL-6, IL-8 and TNF-α, and mitogen-activated protein kinase pathway activation). Pre-incubation with FM-CBA L74 resulted in an inhibition of epithelial barrier damage and inflammation mediated by mitogen-activated protein kinase pathway activation induced by RV infection. Modulating several protective mechanisms, the postbiotic FM-CBAL74 exerted a preventive action against RV infection. This approach could be a disrupting nutritional strategy against one of the most common killers for the pediatric age.
Collapse
Affiliation(s)
- Cristina Bruno
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Laura Pisapia
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Alessia Romano
- ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maddalena Cortese
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Erika Punzo
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy. .,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy. .,European Laboratory for the Investigation of Food-Induced Diseases, University of Naples Federico II, Naples, Italy. .,Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy. .,Task Force for Nutraceuticals and Functional Foods, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
14
|
Kamble N, Gurung A, Kaufer BB, Pathan AA, Behboudi S. Marek's Disease Virus Modulates T Cell Proliferation via Activation of Cyclooxygenase 2-Dependent Prostaglandin E2. Front Immunol 2022; 12:801781. [PMID: 35003129 PMCID: PMC8727754 DOI: 10.3389/fimmu.2021.801781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/06/2021] [Indexed: 02/02/2023] Open
Abstract
Marek’s disease virus (MDV), an avian alphaherpesvirus, infects chickens, transforms CD4+ T cells, and induces immunosuppression early during infection. However, the exact mechanisms involved in MDV-induced immunosuppression are yet to be identified. Here, our results demonstrate that MDV infection in vitro and in vivo induces activation of cyclooxygenase-2 (COX-2) and production of prostaglandin E2 (PGE2). This exerts its inhibitory effects on T cell proliferation at day 21 post infection via PGE2 receptor 2 (EP2) and receptor 4 (EP4). Impairment of the MDV-induced T cell proliferation was associated with downregulation of IL-2 and transferrin uptake in a COX-2/PGE2 dependent manner in vitro. Interestingly, oral administration of a COX-2 inhibitor, meloxicam, during MDV infection inhibited COX-2 activation and rescued T cell proliferation at day 21 post infection. Taken together, our results reveal a novel mechanism that contributes to immunosuppression in the MDV-infected chickens.
Collapse
Affiliation(s)
| | - Angila Gurung
- The Pirbright Institute, Woking, United Kingdom.,Department of Life Sciences, College of Health and Life Sciences, Brunel University, London, United Kingdom
| | | | - Ansar Ahmed Pathan
- Department of Life Sciences, College of Health and Life Sciences, Brunel University, London, United Kingdom
| | - Shahriar Behboudi
- The Pirbright Institute, Woking, United Kingdom.,Faculty of Health and Medical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
15
|
Elean M, Albarracin L, Fukuyama K, Zhou B, Tomokiyo M, Kitahara S, Araki S, Suda Y, Saavedra L, Villena J, Hebert EM, Kitazawa H. Lactobacillus delbrueckii CRL 581 Differentially Modulates TLR3-Triggered Antiviral Innate Immune Response in Intestinal Epithelial Cells and Macrophages. Microorganisms 2021; 9:microorganisms9122449. [PMID: 34946051 PMCID: PMC8704909 DOI: 10.3390/microorganisms9122449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
Lactobacillus delbrueckii subsp. lactis CRL 581 beneficially modulates the intestinal antiviral innate immune response triggered by the Toll-like receptor 3 (TLR3) agonist poly(I:C) in vivo. This study aimed to characterize further the immunomodulatory properties of the technologically relevant starter culture L. delbrueckii subsp. lactis CRL 581 by evaluating its interaction with intestinal epithelial cells and macrophages in the context of innate immune responses triggered by TLR3. Our results showed that the CRL 581 strain was able to adhere to porcine intestinal epithelial (PIE) cells and mucins. The CRL 581 strain also augmented the expression of antiviral factors (IFN-α, IFN-β, Mx1, OAS1, and OAS2) and reduced inflammatory cytokines in PIE cells triggered by TLR3 stimulation. In addition, the influence of L. delbrueckii subsp. lactis CRL 581 on the response of murine RAW macrophages to the activation of TLR3 was evaluated. The CRL 581 strain was capable of enhancing the expression of IFN-α, IFN-β, IFN-γ, Mx1, OAS1, TNF-α, and IL-1β. Of note, the CRL 581 strain also augmented the expression of IL-10 in macrophages. The results of this study show that the high proteolytic strain L. delbrueckii spp. lactis CRL 581 was able to beneficially modulate the intestinal innate antiviral immune response by regulating the response of both epithelial cells and macrophages relative to TLR3 activation.
Collapse
Affiliation(s)
- Mariano Elean
- Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (L.A.); (L.S.)
| | - Leonardo Albarracin
- Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (L.A.); (L.S.)
| | - Kohtaro Fukuyama
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
| | - Binghui Zhou
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
- International Education and Research Center for Food Agricultural Immunology (CFAI), Livestock Immunology Unit, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Mikado Tomokiyo
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
- International Education and Research Center for Food Agricultural Immunology (CFAI), Livestock Immunology Unit, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Shugo Kitahara
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
| | - Shota Araki
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 980-8572, Japan;
| | - Lucila Saavedra
- Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (L.A.); (L.S.)
| | - Julio Villena
- Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (L.A.); (L.S.)
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
- Correspondence: (J.V.); (E.M.H.); (H.K.)
| | - Elvira M. Hebert
- Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (M.E.); (L.A.); (L.S.)
- Correspondence: (J.V.); (E.M.H.); (H.K.)
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (K.F.); (B.Z.); (M.T.); (S.K.); (S.A.)
- International Education and Research Center for Food Agricultural Immunology (CFAI), Livestock Immunology Unit, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: (J.V.); (E.M.H.); (H.K.)
| |
Collapse
|
16
|
Indo Y, Kitahara S, Tomokiyo M, Araki S, Islam MA, Zhou B, Albarracin L, Miyazaki A, Ikeda-Ohtsubo W, Nochi T, Takenouchi T, Uenishi H, Aso H, Takahashi H, Kurata S, Villena J, Kitazawa H. Ligilactobacillus salivarius Strains Isolated From the Porcine Gut Modulate Innate Immune Responses in Epithelial Cells and Improve Protection Against Intestinal Viral-Bacterial Superinfection. Front Immunol 2021; 12:652923. [PMID: 34163470 PMCID: PMC8215365 DOI: 10.3389/fimmu.2021.652923] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
Previously, we constructed a library of Ligilactobacillus salivarius strains from the intestine of wakame-fed pigs and reported a strain-dependent capacity to modulate IFN-β expression in porcine intestinal epithelial (PIE) cells. In this work, we further characterized the immunomodulatory activities of L. salivarius strains from wakame-fed pigs by evaluating their ability to modulate TLR3- and TLR4-mediated innate immune responses in PIE cells. Two strains with a remarkable immunomodulatory potential were selected: L. salivarius FFIG35 and FFIG58. Both strains improved IFN-β, IFN-λ and antiviral factors expression in PIE cells after TLR3 activation, which correlated with an enhanced resistance to rotavirus infection. Moreover, a model of enterotoxigenic E. coli (ETEC)/rotavirus superinfection in PIE cells was developed. Cells were more susceptible to rotavirus infection when the challenge occurred in conjunction with ETEC compared to the virus alone. However, L. salivarius FFIG35 and FFIG58 maintained their ability to enhance IFN-β, IFN-λ and antiviral factors expression in PIE cells, and to reduce rotavirus replication in the context of superinfection. We also demonstrated that FFIG35 and FFIG58 strains regulated the immune response of PIE cells to rotavirus challenge or ETEC/rotavirus superinfection through the modulation of negative regulators of the TLR signaling pathway. In vivo studies performed in mice models confirmed the ability of L. salivarius FFIG58 to beneficially modulate the innate immune response and protect against ETEC infection. The results of this work contribute to the understanding of beneficial lactobacilli interactions with epithelial cells and allow us to hypothesize that the FFIG35 or FFIG58 strains could be used for the development of highly efficient functional feed to improve immune health status and reduce the severity of intestinal infections and superinfections in weaned piglets.
Collapse
Affiliation(s)
- Yuhki Indo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shugo Kitahara
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Mikado Tomokiyo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shota Araki
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Md. Aminul Islam
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Binghui Zhou
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Leonardo Albarracin
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Scientific Computing Laboratory, Computer Science Department, Faculty of Exact Sciences and Technology, National University of Tucuman, Tucuman, Argentina
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli, (CERELA-CONICET), Tucuman, Argentina
| | - Ayako Miyazaki
- Viral Diseases and Epidemiology Research Division, National Institute of Animal Health, NARO, Tsukuba, Japan
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomonori Nochi
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takato Takenouchi
- Animal Bioregulation Unit, Division of Animal Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Hirohide Uenishi
- Animal Bioregulation Unit, Division of Animal Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Hisashi Aso
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Animal Health Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hideki Takahashi
- Laboratory of Plant Pathology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Plant Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Laboratory of Molecular Genetics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Julio Villena
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli, (CERELA-CONICET), Tucuman, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
17
|
Li J, Wang S, Duan J, Le P, Li C, Ding Y, Wang R, Gao Y. The protective mechanism of resveratrol against hepatic injury induced by iron overload in mice. Toxicol Appl Pharmacol 2021; 424:115596. [PMID: 34044072 DOI: 10.1016/j.taap.2021.115596] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Excessive iron deposition can produce toxicity. Liver, as the main storage site of iron, is more vulnerable to excessive iron than other organs. Many studies have found that Resveratrol (RES) can effectively eliminate oxygen free radicals and resist lipid peroxide damage. However, studies investigating the mechanism of how RES prevents liver injury induced by iron overload are few. This study aims to observe the protective effect of RES on liver injury induced by iron overload in mice. Mice, except for the control group, received an intraperitoneal injection of iron dextran (50 mg/kg) every morning. The L-RES and H-RES groups received intragastric administration of low- and high-concentration RES solutions (20 or 50 mg/kg). The deferoxamine (DFO) group was intraperitoneally injected with DFO (50 mg/kg), while the control and iron overload groups were intraperitoneally injected with the same amount of normal saline every afternoon. Two weeks after continuous administration, iron-overloaded mice treated with high and low doses of RES significantly improved liver injury (GOT and GPT) and decreased LDH activity and MDA content and increased SOD and GSH activities (P < 0.01). Morphological tests showed that RES treatment can reduce liver iron deposition and improve liver pathological changes in iron-overloaded mice. Furthermore, RES treatment caused a significant decrease in Ft expression (P < 0.01). In conclusion, RES can alleviate liver injury in iron-overloaded mice. The mechanism may be related to improve the antioxidant capacity and reduce excess iron in the liver.
Collapse
Affiliation(s)
- Jinghan Li
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Simeng Wang
- College of Nursing, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jiaqi Duan
- College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Peixin Le
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Chao Li
- College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yongpei Ding
- College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Rui Wang
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| | - Yonggang Gao
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
18
|
Abstract
Group A rotaviruses (RVAs) are the major cause of severe acute gastroenteritis (AGE) in children under 5 years of age, annually resulting in nearly 130,000 deaths worldwide. Social conditions in developing countries that contribute to decreased oral rehydration and vaccine efficacy and the lack of approved antiviral drugs position RVA as a global health concern. In this minireview, we present an update in the field of antiviral compounds, mainly in relation to the latest findings in RVA virion structure and the viral replication cycle. In turn, we attempt to provide a perspective on the possible treatments for RVA-associated AGE, with special focus on novel approaches, such as those representing broad-spectrum therapeutic options. In this context, the modulation of host factors, lipid droplets, and the viral polymerase, which is highly conserved among AGE-causing viruses, are analyzed as possible drug targets.
Collapse
|
19
|
Cell surface heat shock protein-mediated entry of tumor cell-adapted rotavirus into U-937 cells. Folia Microbiol (Praha) 2021; 66:623-638. [PMID: 33950511 DOI: 10.1007/s12223-020-00845-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/21/2020] [Indexed: 10/21/2022]
Abstract
Rotaviruses infect cells by binding to specific cell surface molecules including gangliosides, heat shock protein cognate protein 70 (Hsc70), and some integrins. The characterization of cell surface receptors defining viral tropism is crucial for inhibiting entry into the normal cells or the cancer cells. In the present work, several tumor cell-adapted rotavirus isolates were tested for their interaction with some heat shock proteins (HSPs) present in the U-937 cells, derived from a human pleural effusion (histiocytic lymphoma monocyte). This interaction was examined by virus overlay protein-binding (VOPB), immunochemistry, immuno-dot blot assays, and flow cytometry. The results indicated that the rotavirus isolates studied were able to infect U937 cells by interacting with Hsp90, Hsp70, Hsp60, Hsp40, Hsc70, protein disulfide isomerase (PDI), and integrin β3, which are implicated in cellular proliferation, differentiation, and cancer development. Interestingly, these cellular proteins were found to be associated in lipid microdomains (rafts), facilitating in this way eventual sequential interactions of the rotavirus particles with the cell surface receptors. The rotavirus tropism for U937 cells through the use of these cell surface proteins made this rotavirus isolates an attractive target for the development of oncolytic strategies in the context of alternative and complementary treatment of cancer.
Collapse
|
20
|
Sander WJ, Fourie C, Sabiu S, O'Neill FH, Pohl CH, O'Neill HG. Reactive oxygen species as potential antiviral targets. Rev Med Virol 2021; 32:e2240. [PMID: 33949029 DOI: 10.1002/rmv.2240] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are by-products of cellular metabolism and can be either beneficial, at low levels, or deleterious, at high levels, to the cell. It is known that several viral infections can increase oxidative stress, which is mainly facilitated by viral-induced imbalances in the antioxidant defence mechanisms of the cell. While the exact role of ROS in certain viral infections (adenovirus and dengue virus) remains unknown, other viruses can use ROS for enhancement of pathogenesis (SARS coronavirus and rabies virus) or replication (rhinovirus, West Nile virus and vesicular stomatitis virus) or both (hepatitis C virus, human immunodeficiency virus and influenza virus). While several viral proteins (mainly for hepatitis C and human immunodeficiency virus) have been identified to play a role in ROS formation, most mediators of viral ROS modulation are yet to be elucidated. Treatment of viral infections, including hepatitis C virus, human immunodeficiency virus and influenza virus, with ROS inhibitors has shown a decrease in both pathogenesis and viral replication both in vitro and in animal models. Clinical studies indicating the potential for targeting ROS-producing pathways as possible broad-spectrum antiviral targets should be evaluated in randomized controlled trials.
Collapse
Affiliation(s)
- Willem J Sander
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Corinne Fourie
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Saheed Sabiu
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa.,Department of Biotechnology and Food Science, Durban University of Technology, Durban, South Africa
| | - Frans H O'Neill
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Carolina H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Hester G O'Neill
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
21
|
Hellysaz A, Hagbom M. Understanding the Central Nervous System Symptoms of Rotavirus: A Qualitative Review. Viruses 2021; 13:v13040658. [PMID: 33920421 PMCID: PMC8069368 DOI: 10.3390/v13040658] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 01/08/2023] Open
Abstract
This qualitative review on rotavirus infection and its complications in the central nervous system (CNS) aims to understand the gut–brain mechanisms that give rise to CNS driven symptoms such as vomiting, fever, feelings of sickness, convulsions, encephalitis, and encephalopathy. There is substantial evidence to indicate the involvement of the gut–brain axis in symptoms such as vomiting and diarrhea. The underlying mechanisms are, however, not rotavirus specific, they represent evolutionarily conserved survival mechanisms for protection against pathogen entry and invasion. The reviewed studies show that rotavirus can exert effects on the CNS trough nervous gut–brain communication, via the release of mediators, such as the rotavirus enterotoxin NSP4, which stimulates neighboring enterochromaffin cells in the intestine to release serotonin and activate both enteric neurons and vagal afferents to the brain. Another route to CNS effects is presented through systemic spread via lymphatic pathways, and there are indications that rotavirus RNA can, in some cases where the blood brain barrier is weakened, enter the brain and have direct CNS effects. CNS effects can also be induced indirectly as a consequence of systemic elevation of toxins, cytokines, and/or other messenger molecules. Nevertheless, there is still no definitive or consistent evidence for the underlying mechanisms of rotavirus-induced CNS complications and more in-depth studies are required in the future.
Collapse
|
22
|
Wedekind SIS, Shenker NS. Antiviral Properties of Human Milk. Microorganisms 2021; 9:715. [PMID: 33807146 PMCID: PMC8066736 DOI: 10.3390/microorganisms9040715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 12/29/2022] Open
Abstract
Humans have always coexisted with viruses, with both positive and negative consequences. Evolutionary pressure on mammals has selected intrinsic properties of lactation and milk to support the relatively immunocompromised neonate from environmental pathogens, as well as support the normal development of diverse immune responses. Human milk supports both adaptive and innate immunity, with specific constituents that drive immune learning and maturation, and direct protection against microorganisms. Viruses constitute one of the most ancient pressures on human evolution, and yet there is a lack of awareness by both public and healthcare professionals of the complexity of human milk as an adaptive response beyond the production of maternal antibodies. This review identifies and describes the specific antiviral properties of human milk and describes how maternal support of infants through lactation is protective beyond antibodies.
Collapse
Affiliation(s)
| | - Natalie S. Shenker
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK;
- Human Milk Foundation, Daniel Hall Building, Rothamsted Institute, Harpenden AL5 2JQ, UK
| |
Collapse
|
23
|
Perez C, Rico J, Guerrero C, Acosta O. Role of heat-shock proteins in infection of human adenocarcinoma cell line MCF-7 by tumor-adapted rotavirus isolates. COLOMBIA MEDICA (CALI, COLOMBIA) 2021; 52:e2024196. [PMID: 33911319 PMCID: PMC8054709 DOI: 10.25100/cm.v51i4.4196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background: Viruses are being used as alternative and complementary tools for treating cancers. Oncolytic viruses exhibit tumor tropism, ability to enhance anti-tumor immunity and ability to be used in combination with conventional chemotherapy and radiotherapy. We have recently selected some rotavirus isolates which are adapted to efficiently infect and kill tumor cell lines. Aim: We tested five tumor cell-adapted rotavirus isolates for their ability to infect the human adenocarcinoma cell line MCF-7. Methods: Cell surface membrane-associated proteins mediating virus particle attachment were characterized using ELISA, immunoprecipitation, FACS analysis, and antibody blocking. Results: It was found that heat shock proteins (HSPs) such as Hsp90, Hsp70, Hsp60, and Hsp40 are expressed on the cell surface forming complexes with protein disulfide isomerase (PDI), integrin β3, and heat shock cognate protein 70 (Hsc70) in lipid raft microdomains. Interaction of rotavirus isolates with these cellular proteins was further confirmed by a competition assay and an inhibition assay involving the HSPs tested. Conclusion: Our findings suggest that the tumor cell-adapted rotavirus isolates studied here offer a promising tool for killing tumor cells, thus encouraging further research into this topic, including animal models.
Collapse
Affiliation(s)
- Claudia Perez
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| | - José Rico
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| | - Carlos Guerrero
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| | - Orlando Acosta
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia. Universidad Nacional de Colombia Universidad Nacional de Colombia Faculty of Medicine Department of Physiological Sciences Bogota Colombia
| |
Collapse
|
24
|
Akhter J, Quéromès G, Pillai K, Kepenekian V, Badar S, Mekkawy AH, Frobert E, Valle SJ, Morris DL. The Combination of Bromelain and Acetylcysteine (BromAc) Synergistically Inactivates SARS-CoV-2. Viruses 2021; 13:v13030425. [PMID: 33800932 PMCID: PMC7999995 DOI: 10.3390/v13030425] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection is the cause of a worldwide pandemic, currently with limited therapeutic options. The spike glycoprotein and envelope protein of SARS-CoV-2, containing disulfide bridges for stabilization, represent an attractive target as they are essential for binding to the ACE2 receptor in host cells present in the nasal mucosa. Bromelain and Acetylcysteine (BromAc) has synergistic action against glycoproteins by breakage of glycosidic linkages and disulfide bonds. We sought to determine the effect of BromAc on the spike and envelope proteins and its potential to reduce infectivity in host cells. Recombinant spike and envelope SARS-CoV-2 proteins were disrupted by BromAc. Spike and envelope protein disulfide bonds were reduced by Acetylcysteine. In in vitro whole virus culture of both wild-type and spike mutants, SARS-CoV-2 demonstrated a concentration-dependent inactivation from BromAc treatment but not from single agents. Clinical testing through nasal administration in patients with early SARS-CoV-2 infection is imminent.
Collapse
Affiliation(s)
- Javed Akhter
- Department of Surgery, St. George Hospital, Sydney, NSW 2217, Australia; (J.A.); (V.K.); (S.B.); (A.H.M.); (S.J.V.)
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia;
| | - Grégory Quéromès
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (G.Q.); (E.F.)
| | | | - Vahan Kepenekian
- Department of Surgery, St. George Hospital, Sydney, NSW 2217, Australia; (J.A.); (V.K.); (S.B.); (A.H.M.); (S.J.V.)
- Hospices Civils de Lyon, EMR 3738 (CICLY), Lyon 1 Université, F-69921 Lyon, France
| | - Samina Badar
- Department of Surgery, St. George Hospital, Sydney, NSW 2217, Australia; (J.A.); (V.K.); (S.B.); (A.H.M.); (S.J.V.)
- St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW 2217, Australia
| | - Ahmed H. Mekkawy
- Department of Surgery, St. George Hospital, Sydney, NSW 2217, Australia; (J.A.); (V.K.); (S.B.); (A.H.M.); (S.J.V.)
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia;
- St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW 2217, Australia
| | - Emilie Frobert
- CIRI, Centre International de Recherche en Infectiologie, Team VirPatH, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France; (G.Q.); (E.F.)
- Laboratoire de Virologie, Institut des Agents Infectieux (IAI), Hospices Civils de Lyon, Groupement Hospitalier Nord, F-69004 Lyon, France
| | - Sarah J. Valle
- Department of Surgery, St. George Hospital, Sydney, NSW 2217, Australia; (J.A.); (V.K.); (S.B.); (A.H.M.); (S.J.V.)
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia;
- St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW 2217, Australia
| | - David L. Morris
- Department of Surgery, St. George Hospital, Sydney, NSW 2217, Australia; (J.A.); (V.K.); (S.B.); (A.H.M.); (S.J.V.)
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia;
- St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW 2217, Australia
- Correspondence: ; Tel.: +61-(02)-91132590
| |
Collapse
|
25
|
Pincemail J, Cavalier E, Charlier C, Cheramy–Bien JP, Brevers E, Courtois A, Fadeur M, Meziane S, Goff CL, Misset B, Albert A, Defraigne JO, Rousseau AF. Oxidative Stress Status in COVID-19 Patients Hospitalized in Intensive Care Unit for Severe Pneumonia. A Pilot Study. Antioxidants (Basel) 2021; 10:257. [PMID: 33562403 PMCID: PMC7914603 DOI: 10.3390/antiox10020257] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A key role of oxidative stress has been highlighted in the pathogenesis of COVID-19. However, little has been said about oxidative stress status (OSS) of COVID-19 patients hospitalized in intensive care unit (ICU). MATERIAL AND METHODS Biomarkers of the systemic OSS included antioxidants (9 assays), trace elements (3 assays), inflammation markers (4 assays) and oxidative damage to lipids (3 assays). RESULTS Blood samples were drawn after 9 (7-11) and 41 (39-43) days of ICU stay, respectively in 3 and 6 patients. Vitamin C, thiol proteins, reduced glutathione, γ-tocopherol, β-carotene and PAOT® score were significantly decreased compared to laboratory reference values. Selenium concentration was at the limit of the lower reference value. By contrast, the copper/zinc ratio (as a source of oxidative stress) was higher than reference values in 55% of patients while copper was significantly correlated with lipid peroxides (r = 0.95, p < 0.001). Inflammatory biomarkers (C-reactive protein and myeloperoxidase) were significantly increased when compared to normals. CONCLUSIONS The systemic OSS was strongly altered in critically ill COVID-19 patients as evidenced by increased lipid peroxidation but also by deficits in some antioxidants (vitamin C, glutathione, thiol proteins) and trace elements (selenium).
Collapse
Affiliation(s)
- Joël Pincemail
- Clinical Chemistry, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (E.C.); (E.B.); (C.L.G.)
| | - Etienne Cavalier
- Clinical Chemistry, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (E.C.); (E.B.); (C.L.G.)
| | - Corinne Charlier
- Toxicology Department, CHU of Liège, Sart Tilman, 4000 Liège, Belgium;
| | - Jean-Paul Cheramy–Bien
- Department of Cardiovascular Surgery, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (J.-P.C.-B.); (A.C.); (J.-O.D.)
| | - Eric Brevers
- Clinical Chemistry, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (E.C.); (E.B.); (C.L.G.)
| | - Audrey Courtois
- Department of Cardiovascular Surgery, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (J.-P.C.-B.); (A.C.); (J.-O.D.)
| | - Marjorie Fadeur
- Service of Diabetology, Nutrition and Metabolic Diseases, CHU of Liège, Sart Tilman, 4000 Liège, Belgium;
| | - Smail Meziane
- Institut Européen des Antioxydants, 54000 Nancy, France;
| | - Caroline Le Goff
- Clinical Chemistry, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (E.C.); (E.B.); (C.L.G.)
| | - Benoît Misset
- Intensive Care Department, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (B.M.); (A.-F.R.)
| | - Adelin Albert
- Biostatistics and Medico-economic Information Department, CHU of Liège, Sart Tilman, 4000 Liège, Belgium;
| | - Jean-Olivier Defraigne
- Department of Cardiovascular Surgery, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (J.-P.C.-B.); (A.C.); (J.-O.D.)
| | - Anne-Françoise Rousseau
- Intensive Care Department, CHU of Liège, Sart Tilman, 4000 Liège, Belgium; (B.M.); (A.-F.R.)
| |
Collapse
|
26
|
|
27
|
Huang H, Liao D, Zhou G, Zhu Z, Cui Y, Pu R. Antiviral activities of resveratrol against rotavirus in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 77:153230. [PMID: 32682225 DOI: 10.1016/j.phymed.2020.153230] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/26/2020] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Rotavirus (RV) is the primary causative agent for viral gastroenteritis among infants and young children worldwide. Currently, no clinically approved and effective antiviral drug for the treatment of RV infection is available. PURPOSE We investigated the potential anti-RV activity of resveratrol and underlying mechanisms by which resveratrol acted against RV. METHODS The anti-RV activity of resveratrol in vitro was evaluated using plaque reduction assays. The effects of resveratrol on yield of virion progeny, viral polyprotein expression and genomic RNA synthesis were respectively investigated using enzyme-linked immunosorbent assays, western blotting and qRT-PCR assays. Further, we also measured the antiviral effect of resveratrol by evaluation of antigen clearance and assessment of changes in proinflammatory cytokines/chemokines in RV-infected neonatal mouse model. RESULTS Our results indicated that 20 μM of resveratrol significantly inhibited RV replication in Caco-2 cell line by suppressing RV RNA synthesis, protein expression, viroplasm plaque formation, progeny virion production, and RV-induced cytopathy independent of the different strains and cell lines of RV that we used. Analysis of the effect of time post-addition of resveratrol indicated that its application inhibited early processes in the RV replication cycle. Further study of the underlying mechanism of anti-RV activity indicated that resveratrol inhibited RV replication by suppressing expression of heat-shock protein 90 (HSP90) mRNA and protein, and that the effect occurred in a dose-dependent manner. Overexpression of HSP90 was found to have attenuated the inhibitory effect of resveratrol on RV replication. Interestingly, the application of resveratrol were found to down-regulate the level of inhibition of RV-mediated MEK1/2 and ERK phosphorylation. Using a RV-infected suckling mice model, we found that application of resveratrol significantly lessened the severity of diarrhea, decreased viral titers, and relieved associated symptoms. Levels of mRNA expression of interleukin-2, interleukin-10, tumor necrosis factor-α, interferon-γ, macrophage inflammatory protein 1, and monocyte chemotactic protein-1 were all found to have been sharply reduced in intestinal tissue from mice which had been treated with resveratrol (10 or 20 mg/kg) after RV infection (p < 0.05). CONCLUSION These findings implied that resveratrol exhibits antiviral activity and could be a promising treatment for rotavirus infection.
Collapse
Affiliation(s)
- Haohai Huang
- Department of Clinical Pharmacy, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China; Central Laboratory, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Dan Liao
- Department of Gynaecology, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Guanghui Zhou
- Department of Rehabilitation medicine, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Zhu Zhu
- Sino-American Cancer Research Institute, Guangdong Medical University, Dongguan, Guangdong, China; Scientific Research Platform, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yejia Cui
- Central Laboratory, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China; Department of Laboratory, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Rong Pu
- Department of Laboratory, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
28
|
Silwal P, Kim JK, Kim YJ, Jo EK. Mitochondrial Reactive Oxygen Species: Double-Edged Weapon in Host Defense and Pathological Inflammation During Infection. Front Immunol 2020; 11:1649. [PMID: 32922385 PMCID: PMC7457135 DOI: 10.3389/fimmu.2020.01649] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are inevitable sources for the generation of mitochondrial reactive oxygen species (mtROS) due to their fundamental roles in respiration. mtROS were reported to be bactericidal weapons with an innate effector function during infection. However, the controlled generation of mtROS is vital for the induction of efficient immune responses because excessive production of mtROS with mitochondrial damage leads to sustained inflammation, resulting in pathological outcomes such as sepsis. Here, we discuss the beneficial and detrimental roles of mtROS in the innate immune system during bacterial, viral, and fungal infections. Recent evidence suggests that several pathogens have evolved multiple strategies to modulate mtROS for their own benefit. We are just beginning to understand the mechanisms by which mtROS generation is regulated and how mtROS affect protective and pathological responses during infection. Several agents/small molecules that prevent the uncontrolled production of mtROS are known to be beneficial in the maintenance of tissue homeostasis during sepsis. mtROS-targeted approaches need to be incorporated into preventive and therapeutic strategies against a variety of infections.
Collapse
Affiliation(s)
- Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
29
|
Pérez C, Rico J, A. Guerrero C, Acosta O. Role of heat-shock proteins in infection of human adenocarcinoma cell line MCF-7 by tumor-adapted rotavirus isolates. Colomb Med (Cali) 2020. [DOI: 10.25100/cm.v52i1.4196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background:
Viruses are being used as alternative and complementary tools for treating cancers. Oncolytic viruses exhibit tumor tropism, ability to enhance anti-tumor immunity and ability to be used in combination with conventional chemotherapy and radiotherapy. We have recently selected some rotavirus isolates which are adapted to efficiently infect and kill tumor cell lines.
Aim:
We tested five tumor cell-adapted rotavirus isolates for their ability to infect the human adenocarcinoma cell line MCF-7.
Methods:
Cell surface membrane-associated proteins mediating virus particle attachment were characterized using ELISA, immunoprecipitation, FACS analysis, and antibody blocking.
Results:
It was found that heat shock proteins (HSPs) such as Hsp90, Hsp70, Hsp60, and Hsp40 are expressed on the cell surface forming complexes with protein disulfide isomerase (PDI), integrin β3, and heat shock cognate protein 70 (Hsc70) in lipid raft microdomains. Interaction of rotavirus isolates with these cellular proteins was further confirmed by a competition assay and an inhibition assay involving the HSPs tested.
Conclusion:
Our findings suggest that the tumor cell-adapted rotavirus isolates studied here offer a promising tool for killing tumor cells, thus encouraging further research into this topic, including animal models.
Collapse
Affiliation(s)
- Claudia Pérez
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| | - José Rico
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| | - Carlos A. Guerrero
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| | - Orlando Acosta
- Universidad Nacional de Colombia, Faculty of Medicine, Department of Physiological Sciences, Bogota, D.C., Colombia
| |
Collapse
|
30
|
|
31
|
The redox metabolic pathways function to limit Anaplasma phagocytophilum infection and multiplication while preserving fitness in tick vector cells. Sci Rep 2019; 9:13236. [PMID: 31520000 PMCID: PMC6744499 DOI: 10.1038/s41598-019-49766-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 02/06/2023] Open
Abstract
Aerobic organisms evolved conserved mechanisms controlling the generation of reactive oxygen species (ROS) to maintain redox homeostasis signaling and modulate signal transduction, gene expression and cellular functional responses under physiological conditions. The production of ROS by mitochondria is essential in the oxidative stress associated with different pathologies and in response to pathogen infection. Anaplasma phagocytophilum is an intracellular pathogen transmitted by Ixodes scapularis ticks and causing human granulocytic anaplasmosis. Bacteria multiply in vertebrate neutrophils and infect first tick midgut cells and subsequently hemocytes and salivary glands from where transmission occurs. Previous results demonstrated that A. phagocytophilum does not induce the production of ROS as part of its survival strategy in human neutrophils. However, little is known about the role of ROS during pathogen infection in ticks. In this study, the role of tick oxidative stress during A. phagocytophilum infection was characterized through the function of different pathways involved in ROS production. The results showed that tick cells increase mitochondrial ROS production to limit A. phagocytophilum infection, while pathogen inhibits alternative ROS production pathways and apoptosis to preserve cell fitness and facilitate infection. The inhibition of NADPH oxidase-mediated ROS production by pathogen infection appears to occur in both neutrophils and tick cells, thus supporting that A. phagocytophilum uses common mechanisms for infection of ticks and vertebrate hosts. However, differences in ROS response to A. phagocytophilum infection between human and tick cells may reflect host-specific cell tropism that evolved during pathogen life cycle.
Collapse
|
32
|
Chard AN, Baker KK, Tsai K, Levy K, Sistrunk JR, Chang HH, Freeman MC. Associations between soil-transmitted helminthiasis and viral, bacterial, and protozoal enteroinfections: a cross-sectional study in rural Laos. Parasit Vectors 2019; 12:216. [PMID: 31064387 PMCID: PMC6505259 DOI: 10.1186/s13071-019-3471-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Background Humans are susceptible to over 1400 pathogens. Co-infection by multiple pathogens is common, and can result in a range of neutral, facilitative, or antagonistic interactions within the host. Soil-transmitted helminths (STH) are powerful immunomodulators, but evidence of the effect of STH infection on the direction and magnitude of concurrent enteric microparasite infections is mixed. Methods We collected fecal samples from 891 randomly selected children and adults in rural Laos. Samples were analyzed for 5 STH species, 6 viruses, 9 bacteria, and 5 protozoa using a quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay. We utilized logistic regression, controlling for demographics and household water, sanitation, and hygiene access, to examine the effect of STH infection on concurrent viral, bacterial, and protozoal infection. Results We found that STH infection was associated with lower odds of concurrent viral infection [odds ratio (OR): 0.48, 95% confidence interval (CI): 0.28–0.83], but higher odds of concurrent bacterial infections (OR: 1.81, 95% CI: 1.06–3.07) and concurrent protozoal infections (OR: 1.50, 95% CI: 0.95–2.37). Trends were consistent across STH species. Conclusions The impact of STH on odds of concurrent microparasite co-infection may differ by microparasite taxa, whereby STH infection was negatively associated with viral infections but positively associated with bacterial and protozoal infections. Results suggest that efforts to reduce STH through preventive chemotherapy could have a spillover effect on microparasite infections, though the extent of this impact requires additional study. The associations between STH and concurrent microparasite infection may reflect a reverse effect due to the cross-sectional study design. Additional research is needed to elucidate the exact mechanism of the immunomodulatory effects of STH on concurrent enteric microparasite infection. Electronic supplementary material The online version of this article (10.1186/s13071-019-3471-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna N Chard
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Kelly K Baker
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Kevin Tsai
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Karen Levy
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Jeticia R Sistrunk
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Howard H Chang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA
| | - Matthew C Freeman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, 30322, USA.
| |
Collapse
|
33
|
Plasmatic Soluble Receptor for Advanced Glycation End Products as a New Oxidative Stress Biomarker in Patients with Prosthetic-Joint-Associated Infections? DISEASE MARKERS 2017; 2017:6140896. [PMID: 29386700 PMCID: PMC5745725 DOI: 10.1155/2017/6140896] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/28/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022]
Abstract
Prosthetic joint infection (PJI) is the most common cause of failure of total joint arthroplasty, but a gold standard for PJI diagnosis is still lacking. Advanced glycation end products (AGEs) are proinflammatory molecules inducing intracellular oxidative stress (OS) after binding to their cell membrane receptors (RAGE). The aim of this study was to evaluate plasmatic soluble receptor for advanced glycation end products (sRAGE), as a new OS and infection marker correlating sRAGE to the level of OS and antioxidant defenses, in PJI, in order to explore the possible application of this new biomarker in the early diagnosis of PJI. Plasmatic sRAGE levels (by ELISA assay), plasma antioxidant total defenses (by lag time method), plasma reactive oxygen species (ROS), and thiobarbituric acid reactive substance (TBARS) levels (by colorimetric assay) were evaluated in 11 PJI patients and in 30 matched controls. ROS and TBARS were significantly higher (p < 0.001) while plasma total antioxidant capacity and sRAGE were significantly lower (p < 0.01) in patients with PJI compared to controls. Our results confirm the OS in PJI and show a strong negative correlation between the level of sRAGE and oxidative status, suggesting the plasmatic sRAGE as a potential marker for improving PJI early diagnosis.
Collapse
|
34
|
Zhuang L, Chen LF, Zhang YB, Liu Z, Xiao XH, Tang W, Wang GC, Song WJ, Li YL, Li MM. Watsonianone A from Rhodomyrtus tomentosa Fruit Attenuates Respiratory-Syncytial-Virus-Induced Inflammation In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3481-3489. [PMID: 28436225 DOI: 10.1021/acs.jafc.7b00537] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Respiratory syncytial virus (RSV) is one of the most common respiratory pathogens. Immoderate inflammation plays a great role in causing RSV-induced diseases. In the present study, watsonianone A, isolated from the fruit of Rhodomyrtus tomentosa (Ait.) Hassk, was found to show a good inhibitory effect on RSV-induced NO production, with a half-maximal inhibitory concentration of 37.2 ± 1.6 μM. Enzyme-linked immunosorbent assay and fluorescence quantitative polymerase chain reaction analyses indicated that watsonianone A markedly reduced both mRNA and protein levels of tumor necrosis factor α, interleukin 6, and monocyte chemoattractant protein 1 in RSV-infected RAW264.7 cells. Mechanistically, watsonianone A inhibited nuclear factor κB (NF-κB) activation by suppressing IκBα phosphorylation. Further analysis revealed that watsonianone A activated the thioredoxin system and decreased intracellular reactive oxygen species (ROS) levels, which are closely associated with NF-κB activation in RSV-infected cells. These results reveal that watsonianone A can attenuate RSV-induced inflammation via the suppression of ROS-sensitive inflammatory signaling.
Collapse
Affiliation(s)
- Ling Zhuang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Li-Feng Chen
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Yu-Bo Zhang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Zhong Liu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Xu-Hui Xiao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Wei Tang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Guo-Cai Wang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Wen-Jun Song
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Yao-Lan Li
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Man-Mei Li
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| |
Collapse
|
35
|
Sander WJ, O'Neill HG, Pohl CH. Prostaglandin E 2 As a Modulator of Viral Infections. Front Physiol 2017; 8:89. [PMID: 28261111 PMCID: PMC5306375 DOI: 10.3389/fphys.2017.00089] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/01/2017] [Indexed: 02/06/2023] Open
Abstract
Viral infections are a major cause of infectious diseases worldwide. Inflammation and the immune system are the major host defenses against these viral infection. Prostaglandin E2 (PGE2), an eicosanoid generated by cyclooxygenases, has been shown to modulate inflammation and the immune system by regulating the expression/concentration of cytokines. The effect of PGE2 on viral infection and replication is cell type- and virus-family-dependent. The host immune system can be modulated by PGE2, with regards to immunosuppression, inhibition of nitrogen oxide (NO) production, inhibition of interferon (IFN) and apoptotic pathways, and inhibition of viral receptor expression. Furthermore, PGE2 can play a role in viral infection directly by increasing the production and release of virions, inhibiting viral binding and replication, and/or stimulating viral gene expression. PGE2 may also have a regulatory role in the induction of autoimmunity and in signaling via Toll-like receptors. In this review the known effects of PGE2 on the pathogenesis of various infections caused by herpes simplex virus, rotavirus, influenza A virus and human immunodeficiency virus as well the therapeutic potential of PGE2 are discussed.
Collapse
Affiliation(s)
| | | | - Carolina H. Pohl
- Department of Microbial, Biochemical and Food Biotechnology, University of the Free StateBloemfontein, South Africa
| |
Collapse
|
36
|
Marvin SA. The Immune Response to Astrovirus Infection. Viruses 2016; 9:v9010001. [PMID: 28042824 PMCID: PMC5294970 DOI: 10.3390/v9010001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/20/2016] [Accepted: 12/28/2016] [Indexed: 12/14/2022] Open
Abstract
Astroviruses are one of the leading causes of pediatric gastroenteritis worldwide and are clinically importantly pathogens in the elderly and immunocompromised populations. Although the use of cell culture systems and small animal models have enhanced our understanding of astrovirus infection and pathogenesis, little is known about the immune response to astrovirus infection. Studies from humans and animals suggest that adaptive immunity is important in restricting classic and novel astrovirus infections, while studies from animal models and cell culture systems suggest that an innate immune system plays a role in limiting astrovirus replication. The relative contribution of each arm of the immune system in restricting astrovirus infection remains unknown. This review summarizes our current understanding of the immune response to astrovirus infection and highlights some of the key questions that stem from these studies. A full understanding of the immune response to astrovirus infection is required to be able to treat and control astrovirus-induced gastroenteritis.
Collapse
Affiliation(s)
- Shauna A Marvin
- Biology Department, Drake University, Des Moines, IA 50311, USA.
| |
Collapse
|
37
|
Feng Y, Ma L, Liu L, Hong HG, Zhang X, Guo F, Huang R, Shi M, Li Y, Zhang L, Fu P. Rhabdomyolysis induced AKI via the regulation of endoplasmic reticulum stress and oxidative stress in PTECs. RSC Adv 2016. [DOI: 10.1039/c6ra18865f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mechanism for the role of ER stress and oxidative stress activation in rhabdomyolysis-associated AKI.
Collapse
|