1
|
Zhou R, Huang R, Zhou S, Lu S, Lin H, Qiu J, Ma S, He J. Sorbicillinoid HSL-2 inhibits the infection of influenza A virus via interaction with the PPAR-γ/NF-κB pathway. J Infect Chemother 2024; 30:1295-1308. [PMID: 38942291 DOI: 10.1016/j.jiac.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Drug resistance is an important factor in the fight against influenza A virus (IAV). Natural products offer a rich source of lead compounds for the discovery of novel antiviral drugs. In a previous study, we isolated the sorbicillinoid polyketide HSL-2 from the mycelium of fungus Trichoderma sp. T-4-1. Here, we show that this compound exerts strong antiviral activity against a panel of IAVs. METHODS The immunofluorescence and qRT-PCR assays were used to detect the inhibitory effect of HSL-2 toward the replication of influenza virus and IAV-induced expression of the pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β. RESULTS The results indicated that HSL-2 inhibited influenza virus replication, and it significantly inhibited IAV-induced overexpression of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β through modulating the PPAR-γ/NF-κB pathway. Notably, this effect was decreased when cells were transfected with PPAR-γ siRNA or treated with the PPAR-γ inhibitor T0070907. In addition, HSL-2 was able to attenuate lung inflammatory responses and to improve lung lesions in a mouse model of IAV infection. CONCLUSIONS In this paper, we identified a microbial secondary metabolite, HSL-2, with anti-influenza virus activity. This report is the first to describe the antiviral activity and mechanism of action of HSL-2, and it provides a new strategy for the development of novel anti-influenza virus drugs from natural sources.
Collapse
Affiliation(s)
- Runhong Zhou
- Department of Pharmacy, Shenzhen Children's Hospital, Shenzhen, China; Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Ruifeng Huang
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Shaofen Zhou
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Shengsheng Lu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Haixing Lin
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jingnan Qiu
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Shuaiqi Ma
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Jian He
- Group of Peptides and Natural Products Research, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| |
Collapse
|
2
|
Mandalari G, Pennisi R, Gervasi T, Sciortino MT. Pistacia vera L. as natural source against antimicrobial and antiviral resistance. Front Microbiol 2024; 15:1396514. [PMID: 39011148 PMCID: PMC11246903 DOI: 10.3389/fmicb.2024.1396514] [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: 03/05/2024] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
Increased global research is focused on the development of novel therapeutics to combat antimicrobial and antiviral resistance. Pistachio nuts represent a good source of protein, fiber, monounsaturated fatty acids, minerals, vitamins, and phytochemicals (carotenoids, phenolic acids, flavonoids and anthocyanins). The phytochemicals found in pistachios are structurally diverse compounds with antimicrobial and antiviral potential, demonstrated as individual compounds, extracts and complexed into nanoparticles. Synergistic effects have also been reported in combination with existing drugs. Here we report an overview of the antimicrobial and antiviral potential of pistachio nuts: studies show that Gram-positive bacterial strains, such as Staphylococcus aureus, are the most susceptible amongst bacteria, whereas antiviral effect has been reported against herpes simplex virus 1 (HSV-1). Amongst the known pistachio compounds, zeaxanthin has been shown to affect both HSV-1 attachment penetration of human cells and viral DNA synthesis. These data suggest that pistachio extracts and derivatives could be used for the topical treatment of S. aureus skin infections and ocular herpes infections.
Collapse
Affiliation(s)
- Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| | - Rosamaria Pennisi
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| | - Teresa Gervasi
- Department of Biomedical and Dental Science and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Maria Teresa Sciortino
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| |
Collapse
|
3
|
Yang H, Fan X, Mao X, Yu B, He J, Yan H, Wang J. The protective role of prebiotics and probiotics on diarrhea and gut damage in the rotavirus-infected piglets. J Anim Sci Biotechnol 2024; 15:61. [PMID: 38698473 PMCID: PMC11067158 DOI: 10.1186/s40104-024-01018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/29/2024] [Indexed: 05/05/2024] Open
Abstract
Rotavirus is one of the pathogenic causes that induce diarrhea in young animals, especially piglets, worldwide. However, nowadays, there is no specific drug available to treat the disease, and the related vaccines have no obvious efficiency in some countries. Via analyzing the pathogenesis of rotavirus, it inducing diarrhea is mainly due to disturb enteric nervous system, destroy gut mucosal integrity, induce intracellular electrolyte imbalance, and impair gut microbiota and immunity. Many studies have already proved that prebiotics and probiotics can mitigate the damage and diarrhea induced by rotavirus infection in hosts. Based on these, the current review summarizes and discusses the effects and mechanisms of prebiotics and probiotics on rotavirus-induced diarrhea in piglets. This information will highlight the basis for the swine production utilization of prebiotics and probiotics in the prevention or treatment of rotavirus infection in the future.
Collapse
Affiliation(s)
- Heng Yang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Xiangqi Fan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China.
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| |
Collapse
|
4
|
Zhu X, Zhang C, Liu L, Xu L, Yao L. Senolytic combination of dasatinib and quercetin protects against diabetic kidney disease by activating autophagy to alleviate podocyte dedifferentiation via the Notch pathway. Int J Mol Med 2024; 53:26. [PMID: 38240118 PMCID: PMC10852012 DOI: 10.3892/ijmm.2024.5350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
The senolytics dasatinib and quercetin (DQ) alleviate age‑related disorders. However, limited information is available regarding the effects of DQ on diabetic kidney disease (DKD). The present study aimed to explore the effects of DQ on DKD and its potential molecular mechanism(s). Dasatinib (5 mg/kg) and quercetin (50 mg/kg) were administered to diabetic db/db mice by gavage for 20 weeks. Body weight, urine albumin‑creatinine ratio (ACR), serum creatinine (Scr), and blood urea nitrogen (BUN) were recorded at the indicated time periods. Periodic acid‑Schiff and Masson's staining were performed to assess the histopathological changes of kidney tissues. Immunohistochemical analysis, immunofluorescence and western blotting were performed to evaluate the expression levels of extracellular matrix (ECM) proteins, autophagic and podocyte differentiation‑related proteins. In addition, mouse podocytes were administered with high‑glucose, DQ and 3‑methyladenine (3‑MA), and the expression levels of autophagic and podocyte differentiation‑related proteins were measured. Moreover, following overexpression of the Notch intracellular domain (NICD), the expression levels of NICD, autophagic and podocyte differentiation‑related proteins were further assessed. DQ significantly reduced the body weight, blood glucose, ACR, Scr and BUN levels and improved the histopathological changes induced in diabetic db/db mice. In addition, DQ caused a significant downregulation of the expression levels of the ECM proteins, improved autophagy and induced an upregulation of the expression levels of podocyte differentiation‑related proteins. Administration of 3‑MA to mice significantly reduced podocyte differentiation, and overexpression of NICD could reverse the effects of DQ on autophagy and podocyte differentiation in vitro. The present study suggests that DQ protects against DKD by activation of autophagy to alleviate podocyte dedifferentiation via the Notch pathway.
Collapse
Affiliation(s)
- Xinwang Zhu
- Department of Nephrology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Congxiao Zhang
- Department of Nephrology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Blood Purification Center, The Fourth People's Hospital of Shenyang, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Linlin Liu
- Department of Nephrology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Li Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524003, P.R. China
| | - Li Yao
- Department of Nephrology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
5
|
崔 艺, 王 德, 谢 东, 王 海, 徐 睿, 唐 潇, 张 印. [Efficacy of navel application of Jianpiwenyang Gel for chronic diarrhea of spleen and stomach weakness type: a randomized controlled trial and analysis of the mechanism]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:217-225. [PMID: 38501406 PMCID: PMC10954536 DOI: 10.12122/j.issn.1673-4254.2024.02.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To investigate the efficacy of Jianpiwenyang Gel (SSWYG) for treating chronic diarrhea and explore its therapeutic mechanism. METHODS Eighty patients with chronic diarrhea of spleen and stomach weakness type were randomized into two groups for interventions with lifestyle adjustment and treatment with bifid triple viable capsules (control group, n=40) or naval application with SSWYG (treatment group, n=40) for one week, after which symptoms of chronic diarrhea were evaluated. The Chinese medicine system pharmacology analysis platform (TCMSP), GeneCards, NCBI, OMIM database and GEO database (GSE14841) were used to obtain the active ingredients and target proteins of SSWYG and chronic diarrhea-related targets. The key targets were obtained by topological analysis for Gene Ontology (GO) and KEGG analyses. The affinity and binding characteristics of SSWYG for specific targets were verified by molecular docking using AutoDock software. RESULTS In both groups, gastrointestinal symptom rating scale (GSRS), Bristol Scale and TCM syndrome scores significantly improved after the treatments (P < 0.05), and better effects were observed in the treatment group (P < 0.05). Sixtyeight targets of SSWYG in treating chronic diarrhea were obtained, and 33 most probable ones were screened out by topological analysis. GO and KEGG analyses identified several chronic diarrhea-related pathways including the TNF and IL-17 pathways. Molecular docking study showed good affinity of the core components of SSWYG for the key targets CASP3, JNK, IL1B, IL6, and AKT1. JUN and CASP3 had the lowest binding energy and the highest stable binding energy with multiple major active ingredients of SSWYG. CONCLUSION SSWYG can significantly improve clinical symptoms of chronic diarrhea possibly by regulating the TNF and IL-17 as well as other pathways via CASP3 and JUN, suggesting a complex therapeutic mechanism of SSWYG involving multiple ingredients and targets and coordinated regulation of multiple pathways.
Collapse
Affiliation(s)
- 艺馨 崔
- 中国人民解放军总医院第六医学中心中医医学部,北京 100853Department of Traditional Chinese Medicine of the Sixth Clinical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - 德财 王
- 航天中心医院,北京 100049Second Outpatient Department, Aerospace Center Hospital, Beijing 100049, China
| | - 东晴 谢
- 中国人民解放军总医院第一医学中心超声科,北京 100853Department of Ultrasound of the First Clinical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - 海明 王
- 中国人民解放军总医院第六医学中心中医医学部,北京 100853Department of Traditional Chinese Medicine of the Sixth Clinical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - 睿鑫 徐
- 中国人民解放军总医院第六医学中心中医医学部,北京 100853Department of Traditional Chinese Medicine of the Sixth Clinical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - 潇然 唐
- 中国人民解放军总医院第六医学中心中医医学部,北京 100853Department of Traditional Chinese Medicine of the Sixth Clinical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - 印 张
- 中国人民解放军总医院第六医学中心中医医学部,北京 100853Department of Traditional Chinese Medicine of the Sixth Clinical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
6
|
Mitra S, Datta Chaudhuri R, Sarkar R, Banerjee S, Mukherjee A, Sharma R, Gope A, Kitahara K, Miyoshi SI, Chawla-Sarkar M. Rotavirus rewires host cell metabolic pathways toward glutamine catabolism for effective virus infection. Gut Microbes 2024; 16:2428425. [PMID: 39567865 PMCID: PMC11583611 DOI: 10.1080/19490976.2024.2428425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/14/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Rotavirus (RV) accounts for 19.11% of global diarrheal deaths. Though GAVI assisted vaccine introduction has curtailed RV induced mortality, factors like RV strain diversity, differential infantile gut microbiome, malnutrition, interference from maternal antibodies and other administered vaccines, etc. often compromise vaccine efficacy. Herein emerges the need of antivirals which can be administered adjunct to vaccination to curb the socio-economic burden stemming from frequent RV infection. Cognisance of pathogen-perturbed host cellular physiology has revolutionized translational research and aided precision-based therapy, particularly for viruses, with no metabolic machinery of their own. To date there has been limited exploration of the host cellular metabolome in context of RV infection. In this study, we explored the endometabolomic landscape of human intestinal epithelial cells (HT-29) on RV-SA11 infection. Significant alteration of host cellular metabolic pathways like the nucleotide biosynthesis pathway, alanine, aspartate and glutamate metabolism pathway, the host citric acid cycle was observed in RV-SA11 infection scenario. Detailed study further revealed that RV replication is exclusively dependent on glutamine metabolism for their propagation in host cells. Glutamine metabolism generates glutamate, aspartate, and asparagine which facilitates virus infection. Abrogation of aspartate biogenesis from glutamine by use of Aminooxyacetic acid (AOAA), significantly curbed RV-SA11 infection in-vitro and in-vivo. Overall, the study improves our understanding of host-rotavirus interactome and recognizes host glutamine metabolism pathway as a suitable target for effective therapeutic intervention against RV infection.
Collapse
Affiliation(s)
- Suvrotoa Mitra
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| | - Ratul Datta Chaudhuri
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| | - Rakesh Sarkar
- Department of Plant and Microbial Biology, University of California, Berkeley, USA
| | - Shreya Banerjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| | - Arpita Mukherjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| | - Ranjana Sharma
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| | - Animesh Gope
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| | - Kei Kitahara
- ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Collaborative Research Center of Okayama University for Infectious Diseases in India, Kolkata, India
| | - Shin-Ichi Miyoshi
- ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Collaborative Research Center of Okayama University for Infectious Diseases in India, Kolkata, India
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Research Center for Intestinal Health Science, Okayama University, Okayama, Japan
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases (presently ICMR-NIRBI), Kolkata, West Bengal, India
| |
Collapse
|
7
|
Huang H, Liao D, He B, Pu R, Cui Y, Zhou G. Deoxyshikonin inhibited rotavirus replication by regulating autophagy and oxidative stress through SIRT1/FoxO1/Rab7 axis. Microb Pathog 2023; 178:106065. [PMID: 36907361 DOI: 10.1016/j.micpath.2023.106065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Rotavirus (RV) is a double-stranded RNA virus. RV prevention and treatment remain a major public health problem due to the lack of clinically specific drugs. Deoxyshikonin is a natural compound isolated from the root of Lithospermum erythrorhizon and one of the shikonin derivatives which owns remarkable therapeutic effects on multiple diseases. The purpose of this research was to inquire Deoxyshikonin's role and mechanism in RV infection. METHODS Deoxyshikonin's function in RV was estimated using Cell Counting Kit-8 analysis, cytopathic effect inhibition assay, virus titer determination, quantitative real-time PCR, enzyme linked-immunosorbent assay, Western blot, immunofluorescence, and glutathione levels detection. Also, Deoxyshikonin's mechanism in RV was appraised with Western blot, virus titer determination, and glutathione levels detection. Moreover, Deoxyshikonin's function in RV in vivo was determined using animal models, and diarrhea score analysis. RESULTS Deoxyshikonin owned anti-RV activity and repressed RV replication in Caco-2 cells. Furthermore, Deoxyshikonin reduced autophagy and oxidative stress caused by RV. Mechanistically, Deoxyshikonin induced low protein levels of SIRT1, ac-Foxo1, Rab7, VP6, low levels of RV titers, low autophagy and oxidative stress. SIRT1 overexpression abolished the effects of Deoxyshikonin on RV-treated Caco-2 cells. Meanwhile, in vivo research affirmed that Deoxyshikonin also possessed anti-RV function, and this was reflected in increased survival rate, body weight, GSH levels, and decreased diarrhea score, RV virus antigen, LC-3II/LC3-I. CONCLUSION Deoxyshikonin reduced RV replication through mediating autophagy and oxidative stress via SIRT1/FoxO1/Rab7 pathway.
Collapse
Affiliation(s)
- Haohai Huang
- Medical and Pharmacy Research Laboratory, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China; Department of Clinical Pharmacy, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China.
| | - Dan Liao
- Medical and Pharmacy Research Laboratory, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China; Department of Gynaecology, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Bin He
- Medical and Pharmacy Research Laboratory, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Rong Pu
- Department of Laboratory, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Yejia Cui
- Department of Laboratory, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Guanghui Zhou
- Department of TCM Rehabilitation, SSL Central Hospital of Dongguan, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| |
Collapse
|