1
|
Wang A, Liu G, Zheng L, Wang S. A review: Mechanism and research progress of the effects of Astragalus polysaccharides on obesity. Int J Biol Macromol 2025:143984. [PMID: 40339857 DOI: 10.1016/j.ijbiomac.2025.143984] [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: 12/20/2024] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
As living standards rise, health has become a top concern, and the issue of obesity has drawn extensive attention. Astragalus polysaccharides (APS), the key active component of Astragalus, have emerged as a promising subject in weight-loss research. Recent breakthroughs in APS studies-such as its dual regulatory effects on gut microbiota and metabolic pathways, novel insights into its anti-inflammatory mechanisms via TLR4/NF-κB signaling, and synergistic interactions with other herbal compounds-warrant an updated synthesis of current knowledge. Previous reviews on APS and obesity have predominantly focused on isolated mechanisms (e.g., lipid metabolism or inflammation), yet a comprehensive analysis integrating its multi-target effects, comparative advantages over conventional anti-obesity drugs, and clinical translation challenges remains lacking. This review uniquely consolidates advances in APS research over the past five years, emphasizing its holistic action on inflammation, insulin resistance, hepatic steatosis, and gut dysbiosis. By systematically comparing APS with pharmacological and nutritional interventions, we highlight its potential as a natural, low-toxicity alternative with multi-organ regulatory capabilities. Furthermore, we address critical gaps in bioavailability optimization and clinical validation, providing a roadmap for future research and therapeutic development.
Collapse
Affiliation(s)
- Anna Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410128, China; Department of Cardiology, The First People's Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling 317500, Zhejiang Province, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410128, China.
| | - Lin Zheng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan 410128, China
| | - Shuangshuang Wang
- Department of Cardiology, The First People's Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling 317500, Zhejiang Province, China.
| |
Collapse
|
2
|
Zhao W, Li J, Cai J, Gao J, Hu Y, Dong C. Research Progress on the Antifibrotic Activity of Traditional Chinese Medicine Polysaccharides. Chem Biodivers 2025; 22:e202402012. [PMID: 39563554 DOI: 10.1002/cbdv.202402012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/21/2024]
Abstract
Fibrosis is a pathological process characterized by excessive extracellular matrix (ECM) deposition and proliferation fibrous tissue, a condition associated with various chronic diseases, such as liver cirrhosis, inflammation of the lungs, and myocarditis. Clinical treatment options for fibrotic diseases are currently limited and have poor efficacy. However, recent studies have increasingly demonstrated that polysaccharides exhibit significant antifibrotic activity by modulating cell proliferation and migration, inhibiting inflammation and oxidative stress associated fibrosis and regulating gut microbiota. This review provides an overview of recent advances in polysaccharide research for antifibrosis and offers new perspectives on the treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Wenjing Zhao
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Jieming Li
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Juntao Cai
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Jie Gao
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Yulong Hu
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| | - Chunhong Dong
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Polysaccharide Research Center, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, China
| |
Collapse
|
3
|
Qian Y, Zhang Y, Chen Y, Wang X, Liu L, Yu H, Wu Z, Gui L, He X, Huang L, Luo H, Xu M, Jiang Y, Li F. Linarin attenuates hyperuricemic nephropathy by modulating Nrf2/Keap1 and TLR4/NF-κB signaling pathways: Linarin attenuates hyperuricemic nephropathy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156440. [PMID: 39908585 DOI: 10.1016/j.phymed.2025.156440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Hyperuricemia (HUA) can lead to hyperuricemic nephropathy (HN) as a result of prolonged uric acid (UA) supersaturation, primarily characterized by excessive inflammation and oxidative stress. In clinical practice, the absence of specific drugs for HN treatment necessitates the use of urate-lowering drugs, despite their lack of reno-protective properties. Linarin, the principal pharmacological constituent of Chrysanthemum indicum L. (C. indicum L.), exhibits diverse bioactivities, including anti-inflammatory, antioxidant, and nephroprotective effects. However, there have been no reports on linarin's ability to mitigate HN, and the underlying mechanisms remain unexplored. PURPOSE This study aimed to investigate the mechanisms of linarin on ameliorating HN, with a particular emphasis on oxidative stress and inflammatory pathways. METHODS A HUA mouse model was developed using male ICR mice treated with hypoxanthine and potassium oxonate. Additionally, an adenosine-induced hyperuricemic cell model was established in NRK-52E cells. Following linarin treatment, serum UA levels and renal function parameters were assessed. The expression of proteins associated with UA production and excretion, oxidative stress, inflammation, and apoptosis was evaluated using western blot, immunohistochemical, and immunofluorescence analyses. Furthermore, Nrf2 knockout mice and Nrf2 inhibitor ML385 were utilized to investigate the mechanism of linarin on improving HN. RESULTS Linarin significantly decreased the serum UA levels, inhibited XO activity and regulated UA transporter in the HUA mice. Moreover, linarin reversed the renal index, serum BUN and Cr levels, along with the expression levels of KIM-1, apoptosis-related molecules. Additionally, linarin obviously reduced the levels of TNF-α, IL-1β and IL-6, and alleviated renal inflammatory via suppressing the TLR4, p-NF-κB and p-IκBα levels. Furthermore, linarin was able to reverse the levels of SOD and MDA, and the expression of Nrf2, Keap1, NQO1, and HO-1 to mitigate oxidative stress both in vitro and in vivo. Inhibition of Nrf2 further confirmed that the renoprotective effect of linarin was linked to the activation of Nrf2. CONCLUSION This study is the first to propose linarin as a potential natural compound for alleviating HN by modulating the Nrf2/Keap1 and TLR4/NF-κB signaling pathways, providing a promising strategy for HN.
Collapse
Affiliation(s)
- Yongshuai Qian
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Department of Pharmacy, Zhengzhou Seventh People's Hospital, Zhengzhou, Henan, 450016, China
| | - Yan Zhang
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Yue Chen
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xiaofei Wang
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Li Liu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Huifan Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Institute of Biomedicine, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Zhengkun Wu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Institute of Biomedicine, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Lili Gui
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China; Institute of Biomedicine, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xuelai He
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Linsheng Huang
- Department of Hepatopancreatobiliary Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Han Luo
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Mao Xu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Ying Jiang
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| | - Fei Li
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, 442000, China.
| |
Collapse
|
4
|
Zhou F, Guo Y, Li W, Hu Y, Yang L, Fu S, Bao X, Tong H, Ye Y, Ding Z. Tetrastigma hemsleyanum polysaccharide protects against "two-hit" induced severe pneumonia via TLR4/NF-κB signaling pathway. Int J Biol Macromol 2025; 303:140639. [PMID: 39909274 DOI: 10.1016/j.ijbiomac.2025.140639] [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: 11/10/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Severe pneumonia, frequently accompanied by cytokine storms, stands as a perilous respiratory condition with alarmingly high mortality rates. Tetrastigma hemsleyanum polysaccharide (THP), a pivotal constituent derived from Tetrastigma hemsleyanum Diels et Gilg (TH), has demonstrated efficacy in treating lung inflammation. However, its precise efficacy and underlying mechanisms in the context of severe pneumonia remain elusive. Our research aims to elucidate THP's protective effects in a "two-hit" severe pneumonia model. Our observations indicate that THP administration markedly shields the lungs from injury, reduces pulmonary apoptosis, balances the formation of immune thrombus and alleviates oxidative stress in pneumonia-induced mice. Furthermore, THP significantly decreases the levels of pro-inflammatory cytokines, suggesting its robust anti-inflammatory capabilities. Notably, THP also plays a crucial role in normalizing gut microbiota imbalance, which is vital in the pathogenesis of severe pneumonia. Metabolomic analysis further validates THP's restorative effects on plasma metabolites, indicating its involvement in regulating energy metabolism and immune homeostasis. Mechanistically, THP targets the TLR4/NF-κB signaling pathway, a core mediator of inflammation, thereby dampening the inflammatory cascade. In summary, our findings underscore that THP, through its multifaceted actions targeting inflammation, oxidative stress, immune thrombus formation, gut microbiota regulation, and metabolic modulation, emerges as a promising therapeutic approach for severe pneumonia. This study provides invaluable insights into the potential applications of natural polysaccharides in treating severe pneumonia and highlights the significance of the TLR4/NF-κB pathway in the disease's progression.
Collapse
Affiliation(s)
- Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ying Guo
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wenxuan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yiwen Hu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Liu Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongbin Tong
- Hangzhou HealthBank Medical Laboratory Co., Ltd., Hangzhou, Zhejiang 310053, China
| | - Yujian Ye
- Department of Dermatology, Third People's Hospital of Hangzhou, Hangzhou, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
5
|
Li F, Peng X, Li W. The interaction between various food components and intestinal microbiota improves human health through the gut-X axis: independently or synergistically. Food Funct 2025; 16:2172-2193. [PMID: 39996355 DOI: 10.1039/d4fo04430d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Food contains various components that improve health by affecting the gut microbiota, primarily by modulating its abundance or altering its diversity. Active substances in food have different effects on the gut microbiota when they act alone or in synergy, resulting in varying impacts on health. The bioactive compounds in food exert different effects on various gut microbiota through multiple pathways, thereby delaying or preventing different kinds of disease. The combination of two or more active compounds may have a synergistic effect, which can more effectively alter the gut microbiota and alleviate diseases through the microbiota-gut-organ axis. According to reports, multiple different food components have similar effects, some of which have been shown to have a synergistic effect on the gut microbiota to promote health. However, there is currently no systematic review of its synergistic effects and mechanisms. There may be more compounds with synergistic effects that have not yet been discovered, while their mechanisms of synergy and ways of impacting host health through the gut microbiota deserve further investigation. The purpose of this review is to systematically summarize the effects of different food components on intestinal flora and health, and further analyze the potential synergies between different food components. PubMed and Google Scholar databases were searched in this review.
Collapse
Affiliation(s)
- Fenfa Li
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Wenzhi Li
- Infinitus (China) Company Ltd, Guangzhou 510405, China.
| |
Collapse
|
6
|
Yin M, Peng J, Chen M, Zhan Q, Zhong H. Rosa Roxburghii Tratt Polysaccharides Prevent Alzheimer's Disease-Like Cognitive Dysfunctions and Pathology in Rats by Regulating the Microbiota-Gut-Brain Axis and Oxidative Stress. J Med Food 2025; 28:232-242. [PMID: 40042065 DOI: 10.1089/jmf.2024.k.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
The microbial-gut-brain axis and oxidative stress may be important to the pathogenesis of Alzheimer's disease (AD). Rosa roxburghii Tratt polysaccharides (RRTP) have a strong antioxidant effect and can affect the gut microbiota, and whether it can affect AD is unknown. So, AlCl3 and d-galactose were used to establish AD model rats, and RRTP was used as an intervention treatment. Morris water maze test was used to detect cognitive functions. The hippocampus was used to observe the pathological changes, and the cortex was used to measure antioxidant markers. The stool was collected for 16S rDNA sequencing. Morris water maze test showed that the learning ability and memory level of AD group rats were decreased, and RRTP intervention could mitigate the injury to a certain extent. In the AD group, hematoxylin-eosin staining revealed changes in the morphology of neurons, silver glycine staining revealed neurofibrillary tangles and Congo red staining revealed β-amyloid. RRTP could ameliorate the above changes to some extent. The results of superoxide dismutase, malondialdehyde, and glutathione peroxidase showed that the antioxidant capacity in the RRTP intervention group was significantly higher than that in the AD group. 16S rDNA sequencing results showed that there were differences in the species composition of gut microbiota, and the ratio of Firmicutes to Bacteroidetes in the AD group was decreased. After RRTP intervention, the proportion of Lactobacillus increased. In conclusion, RRTP may prevent AD pathology and cognitive functions in rats to a certain extent through the microbiota-gut-brain axis and oxidative stress.
Collapse
Affiliation(s)
- Mingyue Yin
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Jiangjiang Peng
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Ming Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Qingqing Zhan
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| | - Hui Zhong
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Guizhou Medical University, Guiyang, China
| |
Collapse
|
7
|
Li B, Zhang T, Tan G, Pu Z, Shen Y. Neuroprotective Effects of Astragalus Polysaccharide on Retina Cells and Ganglion Cell Projection in NMDA-Induced Retinal Injury. Curr Eye Res 2025; 50:282-294. [PMID: 39373214 DOI: 10.1080/02713683.2024.2412304] [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: 02/22/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE Astragalus polysaccharide (APS), a water-soluble heteropolysaccharide, possesses immunomodulatory, anti-inflammatory, and cardioprotective properties. This study investigates the neuroprotective potential of APS in a model of N-Methyl-d-aspartic acid (NMDA)-induced retinal neurodegeneration, aiming to explore its potential as a treatment for retinal degenerative diseases. METHODS Retinal function was evaluated using electroretinography (ERG), optomotor reflex (OMR), and flash visual evoked potentials (FVEP). Retinal inflammatory responses were examined through immunohistochemistry, western blotting (WB), and quantitative reverse transcription PCR (qRT-PCR). To assess the integrity of visual projections, an intravitreal injection of adeno-associated virus (AAV) was employed to trace the projections of retinal ganglion cells (RGCs) to the visual centers. RESULTS APS treatment conferred protection to retinal cells, as indicated by ERG and OMR assessments. And APS intervention mitigated NMDA-induced apoptosis, evidenced by a decrease in TUNEL-positive cells. Furthermore, APS treatment attenuated the NMDA-induced reduction in RGC projections to the visual centers, including the superior colliculus and lateral geniculate nucleus, as demonstrated by AAV tracing. CONCLUSIONS Our findings reveal that APS shields the retina from NMDA-induced damage by inhibiting the NF-κB signaling pathway and reduces the detrimental effects of NMDA on RGC projections to the visual centers. These findings propose APS as a potential novel therapeutic agent for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Baige Li
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Tianlu Zhang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Gao Tan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Zeyuan Pu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
- Frontier Science Center for lmmunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
8
|
Wang J, Sun T, Zhang R, Wang T, Li Y. GelMA@APPA microspheres promote chondrocyte regeneration and alleviate osteoarthritis via Fgfr2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156176. [PMID: 39787690 DOI: 10.1016/j.phymed.2024.156176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/09/2024] [Accepted: 10/20/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND In the context of osteoarthritis (OA), a condition marked by joint degeneration, there is a notable absence of efficacious approaches to promote regenerative healing in chondrocytes. Novel therapeutic strategies like nanomicelles-hydrogel microspheres loaded with Astragalus polysaccharide (GelMA@APPA) offer promising avenues for promoting chondrocyte regeneration and mitigating OA progression. METHODS Astragalus polysaccharide (APS) has been shown to induce chondrocyte proliferation and promote cartilage matrix secretion, demonstrating biological activity associated with chondrocyte regeneration. However, the clinical efficacy of APS remains uncertain. Therefore, this investigation validated the beneficial impact of APS on reducing knee joint damage severity induced by destabilization of the medial meniscus (DMM) in mice. The application of bioinformatics analysis and in vitro experimentation revealed that fibroblast growth factor receptor 2 (Fgfr2) in chondrocytes is a key target protein for APS in ameliorating OA-induced cartilage injury, as the deletion of chondrocyte Fgfr2 resulted in the complete loss of the therapeutic effect of APS. To enhance the efficacy of APS, we incorporated APS into nanoparticle-laden hydrogel microspheres to further bolster its potential in chondrocyte regeneration therapy. Subsequently, we developed GelMA@APPA, which exhibited no significant cytotoxic effects on normal chondrocytes in vitro and could be efficiently internalized by chondrocytes. Following subsequent in vitro and in vivo experiments, we affirmed the beneficial effects of GelMA@APPA on OA mice and cartilage cells damaged by OA, as well as its enhancement of the therapeutic effects of APS. RESULTS APS significantly improved knee joint injuries in OA mice. Bioinformatics and in vitro analyses identified Fgfr2 as a critical target protein for APS's regenerative effects. Disruption of Fgfr2 negated APS's benefits. GelMA@APPA demonstrated good biocompatibility, effective internalization by chondrocytes, and enhanced the therapeutic efficacy of APS in experiments conducted both in vitro and in vivo, improving chondrocyte proliferation and reducing apoptosis. CONCLUSIONS This study demonstrates that GelMA@APPA microspheres effectively promote chondrocyte regeneration and OA treatment by activating Fgfr2. These findings suggest a novel therapeutic mechanism for OA and lay the groundwork for future clinical utilization of GelMA@APPA in regenerative medicine.
Collapse
Affiliation(s)
- Jiakai Wang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Tao Sun
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Rong Zhang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China
| | - Tingting Wang
- Department of Gerontology, The First Hospital of China Medical University, Shenyang 110001, PR China.
| | - Yishuo Li
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang 110001, PR China.
| |
Collapse
|
9
|
Luo Y, Zhou S, Zhang X, Lin Y, Liu J, Cheng W, Zeng Y. The role of the microbiota and metabolites in the treatment of pulmonary fibrosis with UC-MSCs: Integrating fecal metabolomics and 16S rDNA analysis. PLoS One 2025; 20:e0313989. [PMID: 39787138 PMCID: PMC11717254 DOI: 10.1371/journal.pone.0313989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/03/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Pulmonary fibrosis (PF) is a chronic and irreversible interstitial lung disease characterized by a lack of effective therapies. Mesenchymal stem cells (MSCs) have garnered significant interest in the realm of lung regeneration due to their abundant availability, ease of isolation, and capacity for expansion. The objective of our study was to investigate the potential therapeutic role of umbilical cord-derived MSCs (UC-MSCs) in the management of PF, with a focus on the alterations in the gut microbiota and its metabolites during the use of UC-MSCs for the treatment of pulmonary fibrosis, as well as the possible mechanisms involved. METHODS Bleomycin injection was utilized to establish a mouse model of lung fibrosis, followed by the application of 16S rDNA sequencing and LC-MS/MS metabolomics to explore the underlying mechanism of UC-MSC treatment for lung fibrosis. Seventy-five mice were allocated into five groups, namely Control, Model, and low/medium/high dose of UC-MSCs groups, and survival metrics, lung morphology, and the levels of the inflammatory cytokines TNF-α, IL-1β, IL-6, and TGF-β1 were subsequently evaluated. Fecal samples from six mice in each of the Control group, Model group, and UC-MSCs-M groups were collected randomly for 16S rDNA sequencing to analyze the gut microbiota and nontargeted metabolomics. RESULTS In comparison to IPF model mice, the three treatment groups exhibited increased survival rates, restored alveolar morphology, and reduced levels of the inflammatory cytokines TNF-α, IL-1β, IL-6, and TGF-β1, confirming the anti-inflammatory properties of UC-MSCs in IPF treatment. The findings from the 16S rDNA assay indicate that UC-MSCs treatment effectively lower α-diversity induced such as Chao 1 and ACE, as well as β-diversity, leading to a decrease in microbiota abundance. The findings from the metabolomics analysis revealed that the metabolites exhibiting notable variances were primarily composed of Lipids and lipid-like molecules, Organoheterocyclic compounds, Organic acids and derivatives, and Benzenoids, indicating the potential of UC-MSCs to exert antifibrotic effects via these metabolic pathways. CONCLUSION Umbilical cord-derived mesenchymal stem cells (UC-MSCs) ameliorate bleomycin-induced pulmonary fibrosis symptoms in mice by exerting anti-inflammatory effects and mitigating pulmonary fibrosis through the modulation of gut microbiota disorders and their metabolism. These findings offer novel insights into the potential mechanisms and clinical utility of stem cell therapy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yukai Luo
- Fujian Key Laboratory of Lung Stem Cells, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Second Clinical Medical School of Fujian Medical University, Quanzhou, Fujian, China
| | - Shuang Zhou
- Fujian Key Laboratory of Lung Stem Cells, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Second Clinical Medical School of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiaojing Zhang
- Fujian Key Laboratory of Lung Stem Cells, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Second Clinical Medical School of Fujian Medical University, Quanzhou, Fujian, China
| | - Yijian Lin
- Fujian Key Laboratory of Lung Stem Cells, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Second Clinical Medical School of Fujian Medical University, Quanzhou, Fujian, China
| | - Jun Liu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Longyan, Longyan, Fujian, China
| | - Wenzhao Cheng
- Fujian Key Laboratory of Lung Stem Cells, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan, Shandong, China
| | - Yiming Zeng
- Fujian Key Laboratory of Lung Stem Cells, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Second Clinical Medical School of Fujian Medical University, Quanzhou, Fujian, China
- Jinan Microecological Biomedicine Shandong Laboratory, Shounuo City Light West Block, Jinan, Shandong, China
- Respiratory Medicine Center of Fujian Province, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
10
|
Yu Y, Fang J, Li Y, Wang X, Zhang J, Wang J, Sun B. The Novel Effect and Potential Mechanism of Lactoferrin on Organ Fibrosis Prevention. Nutrients 2025; 17:197. [PMID: 39796631 PMCID: PMC11723014 DOI: 10.3390/nu17010197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Organ fibrosis is gradually becoming a human health and safety problem, and various organs of the body are likely to develop fibrosis. The ultimate pathological feature of numerous chronic diseases is fibrosis, and few interventions are currently available to specifically target the pathogenesis of fibrosis. The medical detection of organ fibrosis has gradually matured. However, there is currently no effective treatment method for these diseases. Therefore, we need to strive for developing effective and reliable drugs or substances to treat and prevent fibrotic diseases. Lactoferrin (LF) is a multifunctional glycoprotein with many pathological and physiologically active effects, such as antioxidant, anti-inflammatory and antimicrobial effects, and it protects against pathological and physiological conditions in various disease models. This review summarizes the effects and underlying mechanisms of LF in preventing organ fibrosis. As a naturally active substance, LF can be used as a promising and effective drug for the prevention and remission of fibrotic diseases.
Collapse
Affiliation(s)
| | | | | | | | - Jingjie Zhang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, China-Canada Joint Lab of Food Nutrition and Health, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (J.F.); (Y.L.); (X.W.); (B.S.)
| | - Jing Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, China-Canada Joint Lab of Food Nutrition and Health, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (J.F.); (Y.L.); (X.W.); (B.S.)
| | | |
Collapse
|
11
|
Yuan XH, Zhang SF, Hang Y, Shen YH, Zhang SF, Huang WL, Huang JY, Qian YC, Zhang XL, Li QH, Li L. Fuzheng Huayu recipe inhibits bleomycin-induced pulmonary fibrosis in rats by inhibiting M2 polarization of macrophages via the oxidative phosphorylation pathway. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:62-74. [PMID: 38958642 DOI: 10.1080/10286020.2024.2371050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Fuzheng Huayu recipe (FZHYR) is a Chinese patent medicine for the treatment of fibrosis. The effects of FZHYR on pulmonary fibrosis and macrophage polarization were investigated in vitro. FZHYR inhibited pulmonary inflammation and fibrosis and M2 polarization of macrophages in bleomycin-induced pulmonary fibrosis (BPF) of rat model. Differentially expressed genes were screened by high-throughput mRNA sequencing and GSEA showed that oxidative phosphorylation (OXPHOS) was correlated with BPF. FZHYR inhibited expressions of Ndufa2 and Ndufa6 in lung tissues of BPF rats. These findings suggest that OXPHOS pathway serves as a possible target for pulmonary fibrosis therapy by FZHYR.
Collapse
Affiliation(s)
- Xing-Hua Yuan
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Su-Fang Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yu Hang
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Yan-Hua Shen
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Shan-Fang Zhang
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Wei-Ling Huang
- Jing'an District Hospital of Traditional Chinese Medicine, Shanghai 200072, China
| | - Jing-Yi Huang
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Ye-Chang Qian
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Xiu-Lian Zhang
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| | - Qiu-Hong Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Li Li
- Department of Respiratory Disease, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine (Department of Respiratory Diseases, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine), Shanghai 201900, China
| |
Collapse
|
12
|
Yang P, Zhou Q, Zhang Y, Jia M, Li R, Qu Q, Li Z, Feng M, Tian Y, Ren W, Peng X, Shi X. Exploring the Prebiotic Potential of Fermented Astragalus Polysaccharides on Gut Microbiota Regulation In Vitro. Curr Microbiol 2024; 82:52. [PMID: 39709319 DOI: 10.1007/s00284-024-04035-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Astragalus polysaccharides (APS) are known for their prebiotic properties, and fermentation by probiotics is a promising strategy to enhance the prebiotic activity of polysaccharides. In this study, Lactobacillus rhamnosus was used to ferment APS, and response surface methodology was applied to optimize the fermentation parameters. The optimal conditions were determined as follows: 10.28% APS addition, 5.83% inoculum, 35.6 h of fermentation time, and a temperature of 34.6 °C. Additionally, the effects of Fermented Astragalus polysaccharides (FAPS) on human gut microbiota were investigated through in vitro anaerobic incubation. Fecal samples were obtained from 6 healthy volunteers, which were then individually incubated with FAPS. Results demonstrated that FAPS significantly regulated microbial composition and diversity, increasing the abundance of beneficial gut bacteria such as Lactobacillus, E. faecalis, and Brautobacterium, while inhibiting harmful species such as Shigella, Romboutsia, and Clostridium_sensu_stricto_1. Furthermore, FAPS enhanced the production of short-chain fatty acids (SCFAs), which are increasingly recognized to play a role in intestinal homeostasis. These findings suggested that FAPS offers several advantages in terms of increasing beneficial metabolites and regulating gut microbial composition. This study provides valuable insights for expanding the use of plant-derived polysaccharides in the food industry and for developing functional dietary supplements.
Collapse
Affiliation(s)
- Pengshuo Yang
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Qing Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Yingying Zhang
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Mingyue Jia
- School of Life Science, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Runshuang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Qingsong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Zhixun Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Minfang Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Yuting Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Weishuo Ren
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Xinhui Peng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China
| | - Xinyuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan, Beijing, 102488, China.
- Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science andTechnology Commission, Beijing, 100029, China.
| |
Collapse
|
13
|
Li X, Zhu R, Liu Q, Sun H, Sheng H, Zhu L. Effects of traditional Chinese medicine polysaccharides on chronic diseases by modulating gut microbiota: A review. Int J Biol Macromol 2024; 282:136691. [PMID: 39437951 DOI: 10.1016/j.ijbiomac.2024.136691] [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: 11/23/2023] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Intestinal tract is the largest immune system of human body. Gut microbiota (GM) can produce a large number of metabolites, such as short-chain fatty acids and bile acids, which regulate the physiological health of the host and affect the development of disease. In recent years, traditional Chinese medicine (TCM) polysaccharides have attracted extensive attention with multiple biological activities and low toxicity. TCM polysaccharides can promote the growth of intestinal beneficial bacteria and inhibit the growth of harmful bacteria by regulating the structure and function of GM, thus playing a crucial role in preventing or treating chronic diseases such as inflammatory bowel disease (IBD), obesity, type 2 diabetes mellitus (T2DM), liver diseases, cancer, etc. In this paper, the research progress of TCM polysaccharides in the treatment of chronic diseases such as inflammatory bowel disease, obesity, T2DM, liver diseases, cancer, etc. by modulating GM was reviewed. Meanwhile, this review makes an in-depth discussion on the shortcomings of the research of TCM polysaccharides on chronic diseases by modulating GM, and new valuable prospection for the future researches of TCM polysaccharides are proposed, which will provide new ideas for the further study of TCM polysaccharides.
Collapse
Affiliation(s)
- Xinyu Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Riran Zhu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Henglai Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
14
|
Wang L, Qin N, Shi L, Liu R, Zhu T. Gut Microbiota and Tryptophan Metabolism in Pathogenesis of Ischemic Stroke: A Potential Role for Food Homologous Plants. Mol Nutr Food Res 2024; 68:e2400639. [PMID: 39551995 DOI: 10.1002/mnfr.202400639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/15/2024] [Indexed: 11/19/2024]
Abstract
SCOPE The intestinal flora is involved in the maintenance of human health and the development of diseases, and is closely related to the brain. As an essential amino acid, tryptophan (TRP) participates in a variety of physiological functions in the body and affects the growth and health of the human body. TRP catabolites produced by the gut microbiota are important signaling molecules for microbial communities and host-microbe interactions, and play an important role in maintaining health and disease pathogenesis. METHODS AND RESULTS The review first demonstrates the evidence of TRP metabolism in stroke and the relationship between gut microbiota and TRP metabolism. Furthermore, the review reveals that food homologous plants (FHP) bioactive compounds have been shown to regulate various metabolic pathways of the gut microbiota, including the biosynthesis of valine, leucine, isoleucine, and vitamin B6 metabolism. The most notable metabolic alteration is in TRP metabolism. CONCLUSION The interaction between gut microbiota and TRP metabolism offers a plausible explanation for the notable bioactivities of FHP in the treatment of ischemic stroke (IS). This review enhances the comprehension of the underlying mechanisms associated with the bioactivity of FHP on IS.
Collapse
Affiliation(s)
- Lei Wang
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Na Qin
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| | - Liuliu Shi
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Rujuan Liu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Ting Zhu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| |
Collapse
|
15
|
Jia M, Liu Y, Liu J, Meng J, Cao J, Miao L, Zhang H, Zhu Y, Sun M, Yang J. Xuanfei Baidu decoction ameliorates bleomycin-elicited idiopathic pulmonary fibrosis in mice by regulating the lung-gut crosstalk via IFNγ/STAT1/STAT3 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:155997. [PMID: 39312850 DOI: 10.1016/j.phymed.2024.155997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial pneumonia, the available treatment option is limited because the etiology and pathological process are not well understood. Although gut-lung axis reported with an emerging area of host-associated microbiota exist in many chronic lung diseases, the connection between gut-lung microbiota composition with in-site inflammation in IPF development is not yet established. PURPOSE We aimed to address the microbiota and immunity connection, and make it clear how a listed drug, Xuanfei Baidu Decoction (XFBD) affect the lung-gut crosstalk for IPF amelioration, which was previously reported for restoring disrupted lung in IPF and protecting intestinal injury. METHODS Firstly, Micro-CT (μCT) and histopathology were used to check for pathological changes in the lungs and intestines of bleomycin (BLM)-induced IPF mice. Then, Reverse Transcription and Quantitative Real-time PCR (RT-qPCR) and Western blot (WB) assays were employed to detect the integrity of the barrier of lungs and intestines in IPF mice. Subsequently, flow cytometry and 16S rRNA sequencing were used to evaluate the immune and microbial microenvironment of the lungs and intestines. We analyzed the lung-gut microbiota crosstalk for further mechanism exploration. RESULTS Firstly, we revealed that XFBD protected the integrity of the lung and intestinal barriers in the IPF mice, as evidenced by the up-regulation of ZO-1, Claudin-1, Occludin, and VE Cadherin protein expression. Then, we analyzed the changing microbiota and T cell in the gut-lung axis in IPF, and with XFBD, six highly relevant microenvironments were demonstrated that crossing damaged lung-gut barriers and XFBD could reverse these chaotic bacterial and immunity micro-environment, among them Akkermansia was an essential bacteria affecting the expression of systemic IFN-γ downstream STAT1/STAT3 axis was also studied. XFBD prominently up-regulated the production of IFN-γ and p-STAT1 and down-regulated p-STAT3, consequently exerting effects on the lung barrier and gut barrier. Taken together, XFBD ameliorated BLM-induced IPF mice by regulating IFNγ/STAT1/STAT3 axis. CONCLUSION Altogether, our results revealed that XFBD improved the BLM-elicited IPF mice by regulating gut-lung crosstalk via IFN-γ/STAT1/STAT3 axis and provided a new insight of gut-lung crosstalk in IPF, especially the dynamic changes of microorganisms in the damaged lungs needed to pay more attention during IPF therapy.
Collapse
Affiliation(s)
- Mengjie Jia
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yiman Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jia Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Junyu Meng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiazhen Cao
- Changchun University of Chinese Medicine, No. 1035 Boshuo Road, Jingyue National High Tech Industrial Development Zone, Changchun 130117, China
| | - Lin Miao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Mengmeng Sun
- Changchun University of Chinese Medicine, No. 1035 Boshuo Road, Jingyue National High Tech Industrial Development Zone, Changchun 130117, China.
| | - Jian Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
16
|
Hou S, Wang X, Guo J, Han Y, You J, Tian Z, Zheng X, Zheng S, Ling Y, Pei L, Wu E. Triangle correlations of lung microbiome, host physiology and gut microbiome in a rat model of idiopathic pulmonary fibrosis. Sci Rep 2024; 14:28743. [PMID: 39567656 PMCID: PMC11579350 DOI: 10.1038/s41598-024-80023-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
Changes in lung and gut microbial communities have been associated with idiopathic pulmonary fibrosis (IPF). This study aimed to investigate correlations between microbial changes in the lung and gut and host physiological indices in an IPF model, exploring potential mechanisms of the lung-gut axis in IPF pathogenesis. IPF model rats were established via trans-tracheal injection of bleomycin, with assessments of hematological indices, serum cytokines, lung histopathology, and microbiome alterations. Significant differences in microbial structure and composition were observed in the IPF model compared to controls, with 14 lung and 7 gut microbial genera showing significant abundance changes. Further analysis revealed 20 significant correlations between pulmonary and gut genera. Notably, 11 pairs of correlated genera were linked to the same IPF-related physiological indices, such as hydroxyproline, mean corpuscular volume (MCV), and red cell distribution width-standard deviation (RDW-SD). We identified 24 instances where a lung and a gut genus were each associated with the same physiological index, forming "lung genus-index-gut genus" relationships. Mediation analysis showed that indices like hydroxyproline, MCV, and RDW-SD mediated correlations between 10 lung genera (e.g., Cetobacterium, Clostridium XVIII ) and the gut genus Allobaculum. This study first describes gut-lung microbial interactions in pulmonary fibrosis. Mediation analysis suggests pathways underlying "lung genus-host index-gut genus" and "gut genus-host index-lung genus" correlations, thus providing clues to further elucidate the mechanisms of the "gut-lung axis" in IPF pathogenesis.
Collapse
Affiliation(s)
- Sihan Hou
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China
- Institute of Environmental Biology and Life Support Technology, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xueer Wang
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China
| | - Jiarui Guo
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China
| | - Yue Han
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China
| | - Jia You
- Biotherapy Center, The Seventh Medical Center of PLA General Hospital, Beijing, 100081, China
| | - Zhigang Tian
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, No.804 Shenglijie, Xingqing District, Yinchuan, 750004, China
| | - Xiwei Zheng
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, No.804 Shenglijie, Xingqing District, Yinchuan, 750004, China
| | - Siriguleng Zheng
- Department of Information Technology, Polytechnic College, Beijing, China
| | - Yaqing Ling
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China
| | - Lingpeng Pei
- School of Pharmacy, Minzu University of China, Beijing, China.
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China.
| | - Enqi Wu
- School of Pharmacy, Minzu University of China, Beijing, China.
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Minzu University of China, No. 27 Zhongguancun South Avenue, Beijing, 100081, China.
| |
Collapse
|
17
|
Keshavarz Aziziraftar S, Bahrami R, Hashemi D, Shahryari A, Ramezani A, Ashrafian F, Siadat SD. The beneficial effects of Akkermansia muciniphila and its derivatives on pulmonary fibrosis. Biomed Pharmacother 2024; 180:117571. [PMID: 39418965 DOI: 10.1016/j.biopha.2024.117571] [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: 07/29/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive and debilitating respiratory condition characterized by excessive deposition of extracellular matrix proteins and scarring within the lung parenchyma. Despite extensive research, the pathogenesis of PF remains incompletely understood, and effective therapeutic options are limited. Emerging evidence suggests a potential link between gut microbiota dysbiosis and the development of PF, highlighting the gut-lung axis as a promising therapeutic target. Akkermansia muciniphila (A. muciniphila), a mucin-degrading bacterium residing in the gut mucosal layer, has garnered considerable interest due to its immunomodulatory and anti-inflammatory properties. This study investigates the therapeutic potential of live and pasteurized A. muciniphila, as well as its extracellular vesicles (EVs), in mitigating inflammation and fibrosis in a murine model of carbon tetrachloride (CCl4)-induced PF exacerbated by a high-fat diet (HFD). Male C57BL/6 mice were divided into groups receiving either a normal diet or an HFD, with or without CCl4 administration. The mice were then treated with live or pasteurized A. muciniphila, or its EVs. Lung tissue was analyzed for the expression of inflammatory markers and fibrosis markers using real-time PCR and ELISA. Administration of live and pasteurized A. muciniphila, as well as its EVs, significantly downregulated the expression of inflammatory and fibrosis markers in the lung tissue of CCl4-induced PF mice. Furthermore, these treatments ameliorated the increased production of IL-6 and reduced IL-10 levels observed in the HFD and CCl4-treated groups. These findings suggest that A. muciniphila and its derivatives exert protective effects against pulmonary inflammation and fibrosis, potentially through modulation of the gut-lung axis. The study highlights the therapeutic potential of A. muciniphila and its derivatives as novel interventions for the management of PF, warranting further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Shahrbanoo Keshavarz Aziziraftar
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Department of Pathology, University of California San Francisco, San Francisco, US.
| | - Romina Bahrami
- B.S, Department of Microbiology and Microbial Biotech, Shahid Beheshti University, Tehran, Iran.
| | - Danial Hashemi
- B.S, Department of Animal Science and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Arefeh Shahryari
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Centennial College School of Engineering Technology and Applied Science Biotechnology Program Toronto, Ontario, Canada.
| | - Amitis Ramezani
- Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Fatemeh Ashrafian
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Davar Siadat
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
18
|
Ren C, Wang L, Li X, Tang Y, Zhi X, Zhuang M, Chen Q, Gao X, Lv X, Wang C, Wu X, Liu K, Zhao X, Li Y. Elucidating the mechanism of action of Radix Angelica sinensis (Oliv.) Diels and Radix Astragalus mongholicus Bunge ultrafiltration extract on radiation-induced myocardial fibrosis based on network pharmacology and experimental research. Eur J Pharm Sci 2024; 199:106794. [PMID: 38788908 DOI: 10.1016/j.ejps.2024.106794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024]
Abstract
Myocardial fibrosis can induce cardiac dysfunction and remodeling. Great attention has been paid to traditional chinese medicine (TCM) 's effectiveness in treating MF. Radix Angelica sinensis (Oliv.) Diels and Radix Astragalus mongholicus Bunge ultrafiltration extract (RAS-RA), which is a key TCM compound preparation, have high efficacy in regulating inflammation. However, studies on its therapeutic effect on radiation-induced myocardial fibrosis (RIMF) are rare. In this study, RAS-RA had therapeutic efficacy in RIMF and elucidated its mechanism of action. First, we formulated the prediction network that described the relation of RAS-RA with RIMF according to data obtained in different databases. Then, we conducted functional enrichment to investigate the functions and pathways associated with potential RIMF targets for RAS-RA. In vivo experiments were also performed to verify these functions and pathways. Second, small animal ultrasound examinations, H&E staining, Masson staining, transmission electron microscopy, Enzyme-linked immunosorbent assay (ELISA), Western-blotting, Immunohistochemical method and biochemical assays were conducted to investigate the possible key anti-RIMF pathway in RAS-RA. In total, 440 targets were detected in those 21 effective components of RAS-RA; meanwhile, 1,646 RIMF-related disease targets were also discovered. After that, PPI network analysis was conducted to identify 20 key targets based on 215 overlap gene targets. As indicated by the gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) analysis results, inflammation and PI3K/AKT/mTOR pathways might have important effects on the therapeutic effects on RIMF. Molecular docking analysis revealed high binding of effective components to targets (affinity < -6 kcal/mol). Based on experimental verification results, RAS-RA greatly mitigated myocardial fibrosis while recovering the cardiac activity of rats caused by X-rays. According to relevant protein expression profiles, the PI3K/AKT/mTOR pathway was important for anti-fibrosis effect of RAS-RA. Experimental studies showed that RAS-RA improved cardiac function, decreased pathological damage and collagen fiber deposition in cardiac tissues, and improved the mitochondrial structure of the heart of rats. RAS-RA also downregulated TNF-α, IL-6, and IL-1β levels. Additionally, RAS-RA improved the liver and kidney functions and pathological injury of rat kidney and liver tissues, enhanced liver and kidney functions, and protected the liver and kidneys. RAS-RA also increased PI3K, AKT and mTOR protein levels within cardiac tissues and downregulated α-SMA, Collagen I, and Collagen III. The findings of this study suggested that RAS-RA decreased RIMF by suppressing collagen deposition and inflammatory response by inhibiting the PI3K/AKT/mTOR pathway. Thus, RAS-RA was the potential therapeutic agent used to alleviate RIMF.
Collapse
Affiliation(s)
- Chunzhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Lirong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Xiaojing Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Yan Tang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Xiaodong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000 PR China
| | - Mengjie Zhuang
- Xinjiang Medical University School of Basic Medicine, Urumqi 830000 PR China
| | - Qilin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000 PR China
| | - Xinfang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000 PR China
| | - Chunling Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China
| | - Xinke Zhao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000 PR China.
| | - Yingdong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou 730000 PR China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention and Treatment of Chronic Diseases, Lanzhou 730000 PR China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou 730000 PR China.
| |
Collapse
|
19
|
Li C, Cao Y, Peng Y, Ma T, Wu F, Hua Y, Wang X, Bai T, Wei Y, Ji P. Changes in PI3K/AKT and NRF2/HO-1 signaling expression and intestinal microbiota in bleomycin-induced pulmonary fibrosis. Food Chem Toxicol 2024; 190:114796. [PMID: 38852756 DOI: 10.1016/j.fct.2024.114796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/19/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Pulmonary fibrosis is the outcome of the prolonged interstitial pneumonia, characterized by excessive accumulation of fibroblasts and collagen deposition, leading to its development. This study aimed to study the changes in PI3K/AKT and NRF2/HO-1 signaling expression and intestinal microbiota in a rat model of a novel bleomycin-induced pulmonary fibrosis. The findings of our study showed the model was successfully established. The results showed that the alveolar septum in the model was significantly widened and infiltrated by severe inflammatory cells. Alveolar atrophy occurred due to the formation of multiple inflammatory foci. During this period, fibrous tissue was distributed in strips and patches, primarily around the pulmonary interstitium and bronchus. Moreover, lung damage and fibrosis progressively worsened over time. The mRNA expression of HO-1 and NRF2 in the model decreased while the mRNA expression of HIF-1α, VEGF, PI3K and AKT increased. Furthermore, it was observed to decrease the protein expression of E-cad, HO-1 and NRF2, and increase the protein expression of α-SMA and p-AKT. Additionally, this model leaded to an imbalance in the intestinal microbiota. This study demonstrate that the novel pulmonary fibrosis model activates the NRF2/HO-1 pathway and the PI3K/AKT pathway in rat lung tissues, and leading to intestinal barrier disorder.
Collapse
Affiliation(s)
- Chenchen Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yuxia Cao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yousheng Peng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Ting Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Fanlin Wu
- Lanzhou Institute of Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Yongli Hua
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xiuqin Wang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry, Yinchuan, 750002, Ningxia, China
| | - Tong Bai
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yanming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
| | - Peng Ji
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
| |
Collapse
|
20
|
Zhang A, Wang J, Hu Y, Qiu Y, Dong C. Polysaccharides play an anti-fibrotic role by regulating intestinal flora: A review of research progress. Int J Biol Macromol 2024; 271:131982. [PMID: 38724335 DOI: 10.1016/j.ijbiomac.2024.131982] [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: 10/27/2023] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 06/20/2024]
Abstract
Fibrosis is a common pathological process affecting multiple organs. It refers to an increase in fibrous connective tissue and a decrease in parenchymal cells in damaged tissues or organs. This may lead to structural damage and functional decline or even organ failure. The incidence of fibrosis is increasing worldwide, and the need for safe and effective therapeutic drugs and treatments is pivotal. The intestinal tract has a complex network of exchanging information with various tissues in the body. It contains a sizeable microbial community of which the homeostasis and metabolites are closely related to fibrosis. Polysaccharides are a class of biomolecules present in natural products; they have potential value as anti-fibrotic prebiotics. Recently, polysaccharides have been found to improve fibrosis in different organs by decreasing inflammation and modulating the immune function and intestinal microbiota. In this paper, we reviewed the progress made in research concerning polysaccharides and organ fibrosis in relation to the intestinal microbiota from the pathogenesis of fibrosis to the relationship between the intestinal flora and fibrosis. Furthermore, we provide ideas and references for future polysaccharide-drug discovery and strategies for the treatment of fibrosis.
Collapse
Affiliation(s)
- Aoying Zhang
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; College of Medicine, Pingdingshan University, Pingdingshan, Henan 467000, China
| | - Jie Wang
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; College of Medicine, Pingdingshan University, Pingdingshan, Henan 467000, China
| | - Yulong Hu
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Yuanhao Qiu
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; College of Medicine, Pingdingshan University, Pingdingshan, Henan 467000, China.
| | - Chunhong Dong
- Henan Polysaccharide Research Center, Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China.
| |
Collapse
|
21
|
Yang T, Pan Q, Yue R, Liu G, Zhou Y. Daphnetin alleviates silica-induced pulmonary inflammation and fibrosis by regulating the PI3K/AKT1 signaling pathway in mice. Int Immunopharmacol 2024; 133:112004. [PMID: 38613881 DOI: 10.1016/j.intimp.2024.112004] [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: 12/29/2023] [Revised: 03/17/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
Silicosis is a hazardous occupational disease caused by inhalation of silica, characterized by persistent lung inflammation that leads to fibrosis and subsequent lung dysfunction. Moreover, the complex pathophysiology of silicosis, the challenges associated with early detection, and the unfavorable prognosis contribute to the limited availability of treatment options. Daphnetin (DAP), a natural lactone, has demonstrated various pharmacological properties, including anti-inflammatory, anti-fibrotic, and pulmonary protective effects. However, the effects of DAP on silicosis and its molecular mechanisms remain uncover. This study aimed to evaluate the therapeutic effects of DAP against pulmonary inflammation and fibrosis using a silica-induced silicosis mouse model, and investigate the potential mechanisms and targets through network pharmacology, proteomics, molecular docking, and cellular thermal shift assay (CETSA). Here, we found that DAP significantly alleviated silica-induced lung injury in mice with silicosis. The results of H&E staining, Masson staining, and Sirius red staining indicated that DAP effectively reduced the inflammatory response and collagen deposition over a 28-day period following lung exposure to silica. Furthermore, DAP reduced the number of TUNEL-positive cells, increased the expression levels of Bcl-2, and decreased the expression of Bax and cleaved caspase-3 in the mice with silicosis. More importantly, DAP suppressed the expression levels of NLRP3 signaling pathway-related proteins, including NLRP3, ASC, and cleaved caspase-1, thereby inhibiting silica-induced lung inflammation. Further studies demonstrated that DAP possesses the ability to inhibit the epithelial mesenchymal transition (EMT) induced by silica through the inhibition of the TGF-β1/Smad2/3 signaling pathway. The experimental results of proteomic analysis found that the PI3K/AKT1 signaling pathway was the key targets of DAP to alleviate lung injury induced by silica. DAP significantly inhibited the activation of the PI3K/AKT1 signaling pathway induced by silica in lung tissues. The conclusion was also verified by the results of molecular and CETSA. To further verify this conclusion, the activity of PI3K/AKT1 signaling pathway was inhibited in A549 cells using LY294002. When the A549 cells were pretreated with LY294002, the protective effect of DAP on silica-induced injury was lost. In conclusion, the results of this study suggest that DAP alleviates pulmonary inflammation and fibrosis induced by silica by modulating the PI3K/AKT1 signaling pathway, and holds promise as a potentially effective treatment for silicosis.
Collapse
Affiliation(s)
- Tianye Yang
- School of Pharmaceutical Science, Wuhan University, Wuhan 430071, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi 276005, China.
| | - Qian Pan
- Department of Space Physics, Electronic Information School, Hubei Luojia Laboratory, Wuhan University, 430072 Wuhan, China
| | - Rujing Yue
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi 276005, China
| | - Guanghui Liu
- Department of Ophthalmology, Affiliated People's Hospital (Fujian Provincial People's Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China
| | - Yuanyuan Zhou
- School of Pharmaceutical Science, Wuhan University, Wuhan 430071, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. LTD., Linyi 276005, China
| |
Collapse
|
22
|
Wang F, Dai H, Zhou Z, Shan Y, Yu M, Sun J, Sheng L, Huang L, Meng X, You Y, Sheng M. Astragalus polysaccharides augment BMSC homing via SDF-1/CXCR4 modulation: a novel approach to counteract peritoneal mesenchymal transformation and fibrosis. BMC Complement Med Ther 2024; 24:204. [PMID: 38789949 PMCID: PMC11127382 DOI: 10.1186/s12906-024-04483-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
PURPOSE This study aimed to evaluate the potential of astragalus polysaccharide (APS) pretreatment in enhancing the homing and anti-peritoneal fibrosis capabilities of bone marrow mesenchymal stromal cells (BMSCs) and to elucidate the underlying mechanisms. METHODS Forty male Sprague-Dawley rats were allocated into four groups: control, peritoneal dialysis fluid (PDF), PDF + BMSCs, and PDF + APSBMSCs (APS-pre-treated BMSCs). A peritoneal fibrosis model was induced using PDF. Dil-labeled BMSCs were administered intravenously. Post-transplantation, BMSC homing to the peritoneum and pathological alterations were assessed. Stromal cell-derived factor-1 (SDF-1) levels were quantified via enzyme-linked immunosorbent assay (ELISA), while CXCR4 expression in BMSCs was determined using PCR and immunofluorescence. Additionally, a co-culture system involving BMSCs and peritoneal mesothelial cells (PMCs) was established using a Transwell setup to examine the in vitro effects of APS on BMSC migration and therapeutic efficacy, with the CXCR4 inhibitor AMD3100 deployed to dissect the role of the SDF-1/CXCR4 axis and its downstream impacts. RESULTS In vivo and in vitro experiments confirmed that APS pre-treatment notably facilitated the targeted homing of BMSCs to the peritoneal tissue of PDF-treated rats, thereby amplifying their therapeutic impact. PDF exposure markedly increased SDF-1 levels in peritoneal and serum samples, which encouraged the migration of CXCR4-positive BMSCs. Inhibition of the SDF-1/CXCR4 axis through AMD3100 application diminished BMSC migration, consequently attenuating their therapeutic response to peritoneal mesenchyme-to-mesothelial transition (MMT). Furthermore, APS upregulated CXCR4 expression in BMSCs, intensified the activation of the SDF-1/CXCR4 axis's downstream pathways, and partially reversed the AMD3100-induced effects. CONCLUSION APS augments the SDF-1/CXCR4 axis's downstream pathway activation by increasing CXCR4 expression in BMSCs. This action bolsters the targeted homing of BMSCs to the peritoneal tissue and amplifies their suppressive influence on MMT, thereby improving peritoneal fibrosis.
Collapse
Affiliation(s)
- Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziren Zhou
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaohui Meng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yongqing You
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- Medical Experimental Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China.
| |
Collapse
|
23
|
Ren Y, Zhang Y, Cheng Y, Qin H, Zhao H. Genetic liability of gut microbiota for idiopathic pulmonary fibrosis and lung function: a two-sample Mendelian randomization study. Front Cell Infect Microbiol 2024; 14:1348685. [PMID: 38841114 PMCID: PMC11150651 DOI: 10.3389/fcimb.2024.1348685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/10/2024] [Indexed: 06/07/2024] Open
Abstract
Background The microbiota-gut-lung axis has elucidated a potential association between gut microbiota and idiopathic pulmonary fibrosis (IPF). However, there is a paucity of population-level studies with providing robust evidence for establishing causality. This two-sample Mendelian randomization (MR) analysis aimed to investigate the causal relationship between the gut microbiota and IPF as well as lung function. Materials and methods Adhering to Mendel's principle of inheritance, this MR analysis utilized summary-level data from respective genome-wide association studies (GWAS) involving 211 gut microbial taxa, IPF, and lung function indicators such as FEV1, FVC, and FEV1/FVC. A bidirectional two-sample MR design was employed, utilizing multiple MR analysis methods, including inverse variance-weighted (IVW), weighted median, MR-Egger, and weighted mode. Multivariable MR (MVMR) was used to uncover mediating factors connecting the exposure and outcome. Additionally, comprehensive sensitivity analyses were conducted to ensure the robustness of the results. Results The MR results confirmed four taxa were found causally associated with the risk of IPF. Order Bifidobacteriales (OR=0.773, 95% CI: 0.610-0.979, p=0.033), Family Bifidobacteriaceae (OR=0.773, 95% CI: 0.610-0.979, p=0.033), and Genus RuminococcaceaeUCG009 (OR=0.793, 95% CI: 0.652-0.965, p=0.020) exerted protective effects on IPF, while Genus Coprococcus2 (OR=1.349, 95% CI: 1.021-1.783, p=0.035) promote the development of IPF. Several taxa were causally associated with lung function, with those in Class Deltaproteobacteria, Order Desulfovibrionales, Family Desulfovibrionaceae, Class Verrucomicrobiae, Order Verrucomicrobiales and Family Verrucomicrobiaceae being the most prominent beneficial microbiota, while those in Family Lachnospiraceae, Genus Oscillospira, and Genus Parasutterella were associated with impaired lung function. As for the reverse analysis, MR results confirmed the effects of FEV1 and FVC on the increased abundance of six taxa (Phylum Actinobacteria, Class Actinobacteria, Order Bifidobacteriales, Family Bifidobacteriaceae, Genus Bifidobacterium, and Genus Ruminiclostridium9) with a boosted level of evidence. MVMR suggested monounsaturated fatty acids, total fatty acids, saturated fatty acids, and ratio of omega-6 fatty acids to total fatty acids as potential mediating factors in the genetic association between gut microbiota and IPF. Conclusion The current study suggested the casual effects of the specific gut microbes on the risk of IPF and lung function. In turn, lung function also exerted a positive role in some gut microbes. A reasonable dietary intake of lipid substances has a certain protective effect against the occurrence and progression of IPF. This study provides novel insights into the potential role of gut microbiota in IPF and indicates a possible gut microbiota-mediated mechanism for the prevention of IPF.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
- The Second Clinical Mediccal college, Shanxi Medical University, Taiyuan, China
| | - Yao Zhang
- The Second Clinical Mediccal college, Shanxi Medical University, Taiyuan, China
| | - Yanan Cheng
- The Second Clinical Mediccal college, Shanxi Medical University, Taiyuan, China
| | - Hao Qin
- The Second Clinical Mediccal college, Shanxi Medical University, Taiyuan, China
| | - Hui Zhao
- Department of Pulmonary and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
24
|
Yang C, Tan Y, Li Z, Hu L, Chen Y, Zhu S, Hu J, Huai T, Li M, Zhang G, Rao D, Fei G, Shao M, Ding Z. Pulmonary redox imbalance drives early fibroproliferative response in moderate/severe coronavirus disease-19 acute respiratory distress syndrome and impacts long-term lung abnormalities. Ann Intensive Care 2024; 14:72. [PMID: 38735020 PMCID: PMC11089033 DOI: 10.1186/s13613-024-01293-3] [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: 12/09/2023] [Accepted: 04/10/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND COVID-19-associated pulmonary fibrosis remains frequent. This study aimed to investigate pulmonary redox balance in COVID-19 ARDS patients and possible relationship with pulmonary fibrosis and long-term lung abnormalities. METHODS Baseline data, chest CT fibrosis scores, N-terminal peptide of alveolar collagen III (NT-PCP-III), transforming growth factor (TGF)-β1, superoxide dismutase (SOD), reduced glutathione (GSH), oxidized glutathione (GSSG) and malondialdehyde (MDA) in bronchoalveolar lavage fluid (BALF) were first collected and compared between SARS-CoV-2 RNA positive patients with moderate to severe ARDS (n = 65, COVID-19 ARDS) and SARS-CoV-2 RNA negative non-ARDS patients requiring mechanical ventilation (n = 63, non-ARDS). Then, correlations between fibroproliferative (NT-PCP-III and TGF-β1) and redox markers were analyzed within COVID-19 ARDS group, and comparisons between survivor and non-survivor subgroups were performed. Finally, follow-up of COVID-19 ARDS survivors was performed to analyze the relationship between pulmonary abnormalities, fibroproliferative and redox markers 3 months after discharge. RESULTS Compared with non-ARDS group, COVID-19 ARDS group had significantly elevated chest CT fibrosis scores (p < 0.001) and NT-PCP-III (p < 0.001), TGF-β1 (p < 0.001), GSSG (p < 0.001), and MDA (p < 0.001) concentrations on admission, while decreased SOD (p < 0.001) and GSH (p < 0.001) levels were observed in BALF. Both NT-PCP-III and TGF-β1 in BALF from COVID-19 ARDS group were directly correlated with GSSG (p < 0.001) and MDA (p < 0.001) and were inversely correlated with SOD (p < 0.001) and GSH (p < 0.001). Within COVID-19 ARDS group, non-survivors (n = 28) showed significant pulmonary fibroproliferation (p < 0.001) with more severe redox imbalance (p < 0.001) than survivors (n = 37). Furthermore, according to data from COVID-19 ARDS survivor follow-up (n = 37), radiographic residual pulmonary fibrosis and lung function impairment improved 3 months after discharge compared with discharge (p < 0.001) and were associated with early pulmonary fibroproliferation and redox imbalance (p < 0.01). CONCLUSIONS Pulmonary redox imbalance occurring early in COVID-19 ARDS patients drives fibroproliferative response and increases the risk of death. Long-term lung abnormalities post-COVID-19 are associated with early pulmonary fibroproliferation and redox imbalance.
Collapse
Affiliation(s)
- Chun Yang
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yuanyuan Tan
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Zihao Li
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Lei Hu
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yuanyuan Chen
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Shouliang Zhu
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Jiawei Hu
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Tingting Huai
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Mingqing Li
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Guobin Zhang
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China
| | - Dewang Rao
- Anhui Medical University, #81 Meishan Road, Hefei, 230032, Anhui, China
| | - Guanghe Fei
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Min Shao
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Zhenxing Ding
- The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
25
|
Liu B, Zhang X, Liu Z, Pan H, Yang H, Wu Q, Lv Y, Shen T. A novel model for predicting prognosis in patients with idiopathic pulmonary fibrosis based on endoplasmic reticulum stress-related genes. Cell Biol Int 2024; 48:483-495. [PMID: 38238919 DOI: 10.1002/cbin.12121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 03/13/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease of unknown pathogenic origin. Endoplasmic reticulum (ER) stress refers to the process by which cells take measures to ER function when the morphology and function of the reticulum are changed. Recent studies have demonstrated that the ER was involved in the evolution and progression of IPF. In this study, we obtained transcriptome data and relevant clinical information from the Gene Expression Omnibus database and conducted bioinformatics analysis. Among the 544 ER stress-related genes (ERSRGs), 78 were identified as differentially expressed genes (DEGs). These DEGs were primarily enriched in response to ER stress, protein binding, and protein processing. Two genes (HTRA2 and KTN1) were included for constructing an accurate molecular signature. The overall survival of patients was remarkably worse in the high-risk group than in the low-risk group. We further analyzed the difference in immune cells between high-risk and low-risk groups. M0 and M2 macrophages were significantly increased in the high-risk group. Our results suggested that ERSRGs might play a critical role in the development of IPF by regulating the immune microenvironment in the lungs, which provide new insights on predicting the prognosis of patients with IPF.
Collapse
Affiliation(s)
- Bin Liu
- Department of Medical Aspects of Specifc Environments, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Xiang Zhang
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Zikai Liu
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Haihong Pan
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Hongxu Yang
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Qing Wu
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Yan Lv
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Tong Shen
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, China
| |
Collapse
|
26
|
Wen J, Wang C, Song LY, Wang YY, Liang PT, Pang WL, Yin W, Zhang Q, Zhao WT, Sun XP, Yan JY, Yang ZS. Ferroptosis Mediates Pulmonary Fibrosis: Implications for the Effect of Astragalus and Panax notoginseng Decoction. Can Respir J 2024; 2024:5554886. [PMID: 38584671 PMCID: PMC10997418 DOI: 10.1155/2024/5554886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 04/09/2024] Open
Abstract
Objective To investigate the mechanism through which Astragalus and Panax notoginseng decoction (APD) facilitates the treatment of ferroptosis-mediated pulmonary fibrosis. Materials and Methods First, the electromedical measurement systems were used to measure respiratory function in mice; the lungs were then collected for histological staining. Potential pharmacologic targets were predicted via network pharmacology. Finally, tests including immunohistochemistry, reverse transcription-quantitative polymerase chain reaction, and western blotting were used to evaluate the relative expression levels of collagen, transforming growth factor β, α-smooth muscle actin, hydroxyproline, and ferroptosis-related genes (GPX4, SLC7A11, ACSL4, and PTGS2) and candidates involved in the mediation of pathways associated with ferroptosis (Hif-1α and EGFR). Results APD prevented the occurrence of restrictive ventilation dysfunction induced by ferroptosis. Extracellular matrix and collagen fiber deposition were significantly reduced when the APD group compared with the model group; furthermore, ferroptosis was attenuated, expression of PTGS2 and ACSL4 increased, and expression of GPX4 and SLC7A11 decreased. In the APD group, the candidates related to the mediation of ferroptosis (Hif-1α and EGFR) decreased compared with the model group. Discussion and Conclusions. APD may ameliorate restrictive ventilatory dysfunction through the inhibition of ferroptosis. This was achieved through the attenuation of collagen deposition and inflammatory recruitment in pulmonary fibrosis. The underlying mechanisms might involve Hif-1α and EGFR.
Collapse
Affiliation(s)
- Jing Wen
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Cui Wang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Li-yun Song
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yin-ying Wang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Peng-tao Liang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wen-lin Pang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wen Yin
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qiang Zhang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wei-tian Zhao
- Dali Prefectural Hospital of Traditional Chinese Medicine, Dali, Yunnan, China
| | - Xue-ping Sun
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jin-yuan Yan
- Central Laboratory, Kunming Medical University Second Hospital, Kunming, Yunnan, China
| | - Zhong-shan Yang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
27
|
Guo J, Yang L. Regulation effect of the intestinal flora and intervention strategies targeting the intestinal flora in alleviation of pulmonary fibrosis development. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:293-299. [PMID: 39364128 PMCID: PMC11444866 DOI: 10.12938/bmfh.2023-100] [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: 11/24/2023] [Accepted: 02/25/2024] [Indexed: 10/05/2024]
Abstract
Pulmonary fibrosis is an end-stage respiratory disease characterized by fibroblast proliferation and accumulation of extracellular matrix and collagen, which is accompanied by inflammatory damage. The disease is mainly based on pulmonary dysfunction and respiratory failure, the incidence of it is increasing year by year, and the current treatment methods for it are limited. In recent years, it has been found that gut microbes play a crucial role in the pathogenesis and development of pulmonary fibrosis. The microecological disturbance caused by changes in the composition of the intestinal flora can affect the course of pulmonary fibrosis. The regulatory network or information exchange system for gut-lung crosstalk is called the "gut-lung axis". This review focuses on the frontier research on entero-pulmonary regulation in pulmonary fibrosis and on intervention strategies for changing the gut microbiota to improve pulmonary fibrosis, including fecal microbiota transplantation, traditional Chinese medicine interventions, and supplementation with probiotics. In addition, the present problems in this field are also raised in order to provide strong theoretical and strategic support for the future exploration of regulatory mechanisms and therapeutic drug development. This paper reviews the interaction of the intestinal flora with pulmonary fibrosis, introduces the research progress for improving pulmonary fibrosis through interventions targeted at the intestinal flora, and provides new ideas for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jianquan Guo
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi 030001, PR China
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Liyang Yang
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, PR China
| |
Collapse
|
28
|
Liu YX, Song XM, Dan LW, Tang JM, Jiang Y, Deng C, Zhang DD, Li YZ, Wang W. Astragali Radix: comprehensive review of its botany, phytochemistry, pharmacology and clinical application. Arch Pharm Res 2024; 47:165-218. [PMID: 38493280 DOI: 10.1007/s12272-024-01489-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Astragali Radix (A. Radix) is the dried root of Astragalus membranaceus var. mongholicus (Bge) Hsiao or Astragalus membranaceus (Fisch.) Bge., belonging to the family Leguminosae, which is mainly distributed in China. A. Radix has been consumed as a tonic in China for more than 2000 years because of its medicinal effects of invigorating the spleen and replenishing qi. Currently, more than 400 natural compounds have been isolated and identified from A. Radix, mainly including saponins, flavonoids, phenylpropanoids, alkaloids, and others. Modern pharmacological studies have shown that A. Radix has anti-tumor, anti-inflammatory, immunomodulatory, anti-atherosclerotic, cardioprotective, anti-hypertensive, and anti-aging effects. It has been clinically used in the treatment of tumors, cardiovascular diseases, and cerebrovascular complications associated with diabetes with few side effects and high safety. This paper reviewed the progress of research on its chemical constituents, pharmacological effects, clinical applications, developing applications, and toxicology, which provides a basis for the better development and utilization of A. Radix.
Collapse
Affiliation(s)
- Ya-Xiao Liu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Xiao-Mei Song
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Xianyang, 712046, Shaanxi, China
| | - Lin-Wei Dan
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Jia-Mei Tang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Yi Jiang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Xianyang, 712046, Shaanxi, China
| | - Chong Deng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Xianyang, 712046, Shaanxi, China
| | - Dong-Dong Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Xianyang, 712046, Shaanxi, China
| | - Yu-Ze Li
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China.
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Xianyang, 712046, Shaanxi, China.
| | - Wei Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China.
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Xianyang, 712046, Shaanxi, China.
| |
Collapse
|
29
|
Dong Y, He L, Zhu Z, Yang F, Ma Q, Zhang Y, Zhang X, Liu X. The mechanism of gut-lung axis in pulmonary fibrosis. Front Cell Infect Microbiol 2024; 14:1258246. [PMID: 38362497 PMCID: PMC10867257 DOI: 10.3389/fcimb.2024.1258246] [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: 07/13/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Pulmonary fibrosis (PF) is a terminal change of a lung disease that is marked by damage to alveolar epithelial cells, abnormal proliferative transformation of fibroblasts, excessive deposition of extracellular matrix (ECM), and concomitant inflammatory damage. Its characteristics include short median survival, high mortality rate, and limited treatment effectiveness. More in-depth studies on the mechanisms of PF are needed to provide better treatment options. The idea of the gut-lung axis has emerged as a result of comprehensive investigations into the microbiome, metabolome, and immune system. This theory is based on the material basis of microorganisms and their metabolites, while the gut-lung circulatory system and the shared mucosal immune system act as the connectors that facilitate the interplay between the gastrointestinal and respiratory systems. The emergence of a new view of the gut-lung axis is complementary and cross-cutting to the study of the mechanisms involved in PF and provides new ideas for its treatment. This article reviews the mechanisms involved in PF, the gut-lung axis theory, and the correlation between the two. Exploring the gut-lung axis mechanism and treatments related to PF from the perspectives of microorganisms, microbial metabolites, and the immune system. The study of the gut-lung axis and PF is still in its early stages. This review systematically summarizes the mechanisms of PF related to the gut-lung axis, providing ideas for subsequent research and treatment of related mechanisms.
Collapse
Affiliation(s)
- Yawei Dong
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Lanlan He
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhongbo Zhu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Fan Yang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Quan Ma
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanmei Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xuhui Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiping Liu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
30
|
Wang L, Wang J, Yang Z, Wang Y, Zhao T, Luo W, Liang T, Yang Z. Traditional herbs: mechanisms to combat cellular senescence. Aging (Albany NY) 2023; 15:14473-14505. [PMID: 38054830 PMCID: PMC10756111 DOI: 10.18632/aging.205269] [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: 06/30/2023] [Accepted: 10/15/2023] [Indexed: 12/07/2023]
Abstract
Cellular senescence plays a very important role in the ageing of organisms and age-related diseases that increase with age, a process that involves physiological, structural, biochemical and molecular changes in cells. In recent years, it has been found that the active ingredients of herbs and their natural products can prevent and control cellular senescence by affecting telomerase activity, oxidative stress response, autophagy, mitochondrial disorders, DNA damage, inflammatory response, metabolism, intestinal flora, and other factors. In this paper, we review the research information on the prevention and control of cellular senescence in Chinese herbal medicine through computer searches of PubMed, Web of Science, Science Direct and CNKI databases.
Collapse
Affiliation(s)
- Lei Wang
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| | - Zhihui Yang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| | - Yue Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| | - Tiejian Zhao
- Department of Physiology, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| | - Weisheng Luo
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, China
| | - Tianjian Liang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| | - Zheng Yang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi 530222, China
| |
Collapse
|
31
|
Shen Y, Song M, Wu S, Zhao H, Zhang Y. Plant-Based Dietary Fibers and Polysaccharides as Modulators of Gut Microbiota in Intestinal and Lung Inflammation: Current State and Challenges. Nutrients 2023; 15:3321. [PMID: 37571257 PMCID: PMC10420973 DOI: 10.3390/nu15153321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Recent research has underscored the significant role of gut microbiota in managing various diseases, including intestinal and lung inflammation. It is now well established that diet plays a crucial role in shaping the composition of the microbiota, leading to changes in metabolite production. Consequently, dietary interventions have emerged as promising preventive and therapeutic approaches for managing these diseases. Plant-based dietary fibers, particularly polysaccharides and oligosaccharides, have attracted attention as potential therapeutic agents for modulating gut microbiota and alleviating intestinal and lung inflammation. This comprehensive review aims to provide an in-depth overview of the current state of research in this field, emphasizing the challenges and limitations associated with the use of plant-based dietary fibers and polysaccharides in managing intestinal and lung inflammation. By shedding light on existing issues and limitations, this review seeks to stimulate further research and development in this promising area of therapeutic intervention.
Collapse
Affiliation(s)
- Yu Shen
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| | - Mingming Song
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| | - Shihao Wu
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| | - Hongbo Zhao
- College of Rehabilitation Medicine, Jiamusi University, Jiamusi 154007, China
| | - Yu Zhang
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| |
Collapse
|