1
|
Elgendy SA, Soliman MM, Shukry M, Mohammed LA, Nasr HE, Althobaiti S, Almalki DA, Alotaibi KS, Albattal SB, Elnoury HA. Screening impacts of Tilmicosin-induced hepatic and renal toxicity in rats: protection by Rhodiola rosea extract through the involvement of oxidative stress, antioxidants, and inflammatory cytokines biomarkers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7623-7637. [PMID: 38689072 DOI: 10.1007/s00210-024-03089-5] [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: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 05/02/2024]
Abstract
Tilmicosin (TIL) is a semisynthetic macrolide antibiotic with a broad spectrum of activity derived from tylosin. TIL is effective in the treatment of bovine and ovine respiratory diseases caused by different microbes. In parallel, Rhodiola rosea (RHO) is a popular herbal remedy because of its anti-inflammatory and antioxidant qualities. The experiment lasted for 12 days. Depending on the experimental group, the animals received either distilled water or RHO root extract dissolved in distilled water for 12 days through a stomach tube, and the single subcutaneous injection on day 6 of the experiment of either 500 μL of 0.9% NaCl or TIL dissolved in 500 μL 0.9% NaCl. Samples and blood were collected for serum analysis, gene expression, and immunohistochemistry screening at liver and kidney levels. TIL injection increased serum levels of hepatic and renal markers (ALP, ALT, AST, TC, TG, creatinine, and urea) with decreased total proteins. In parallel, TIL induced hepatic and renal oxidative stress as there was an increase in malondialdehyde levels, with a decrease in catalase and reduced glutathione activities. Of interest, pre-administration of RHO inhibited TIL-induced increase in hepato-renal markers, decreased oxidative stress, and increased liver and kidney antioxidant activities. Quantitative RT-PCR showed that TIL increased the liver's HSP70 (heat shock protein), NFkB, and TNF-α mRNA expression. Moreover, TIL upregulated the expression of desmin, nestin, and vimentin expression in the kidney. The upregulated genes were decreased significantly in the protective group that received RHO. Serum inflammatory cytokines and genes of inflammatory markers were affected in liver tissues (HSP70, NFkB, and TNF-α) and kidney tissues (desmin, nestin, and vimentin)-TIL-induced hepatic vacuolation and congestion together with glomerular atrophy. The immunoreactivity of PCNA and HMGB1 was examined immunohistochemically. At cellular levels, PCNA was decreased while HMGB1 immunoreactivity was increased in TIL-injected rats, which was improved by pre-administration of RHO. RHO administration protected the altered changes in liver and renal histology. Current findings support the possible use of RHO to shield the liver and kidney from the negative effects of tilmicosin.
Collapse
Affiliation(s)
- Salwa A Elgendy
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| | - Mohamed Mohamed Soliman
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia.
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Lina Abdelhady Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| | - Hend Elsayed Nasr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| | - Saed Althobaiti
- Department of Biology, Turabah University College, Taif University, Taif, 21995, Saudi Arabia
| | - Daklallah A Almalki
- Biology Department, Faculty of Science and Arts, Al-Mikhwah, Al-Baha University, Al-Baha, Saudi Arabia
| | - Khalid S Alotaibi
- General Science and English Language Department, College of Applied Sciences, AlMaarefa University, Riyadh, Saudi Arabia
| | - Shatha B Albattal
- General Science and English Language Department, College of Applied Sciences, AlMaarefa University, Riyadh, Saudi Arabia
| | - Heba A Elnoury
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, 13511, Egypt
| |
Collapse
|
2
|
Gostyńska A, Buzun K, Żółnowska I, Krajka-Kuźniak V, Mańkowska-Wierzbicka D, Jelińska A, Stawny M. Natural bioactive compounds-The promising candidates for the treatment of intestinal failure-associated liver disease. Clin Nutr 2024; 43:1952-1971. [PMID: 39032247 DOI: 10.1016/j.clnu.2024.07.004] [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: 02/26/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Parenteral nutrition (PN) is a life-saving procedure conducted to maintain a proper nutritional state in patients with severe intestinal failure who cannot be fed orally. A serious complication of PN therapy is liver failure, known as intestinal failure-associated liver disease (IFALD). The pathogenesis of IFALD is multifactorial and includes inhibition of the farnesoid X receptor (FXR) by PN components, bacteria translocation from impaired intestines, and intravenous line-associated bloodstream infection. Currently, the most frequently researched therapeutic option for IFALD is using lipid emulsions based on soy or fish oil and, therefore, free from phytosterols known as FXR antagonists. Nevertheless, the potential side effects of the lack of soybean oil delivery seem to outweigh the benefits, especially in the pediatric population. PN admixture provides all the necessary nutrients; however, it is deprived of exogenous natural bioactive compounds (NBCs) of plant origin, such as polyphenols, characterized by health-promoting properties. Among them, many substances have already been known to demonstrate the hepatoprotective effect in various liver diseases. Therefore, searching for new therapeutic options for IFALD among NBCs seems reasonable and potentially successful. This review summarizes the recent research on polyphenols and their use in treating various liver diseases, especially metabolic dysfunction-associated steatotic liver diseases (MASLD). Furthermore, based on scientific reports, we have described the molecular mechanism of action of selected NBCs that exert hepatoprotective properties. We also summarized the current knowledge on IFALD pathogenesis, described therapeutic options undergoing clinical trials, and presented the future perspective of the potential use of NBCs in PN therapy.
Collapse
Affiliation(s)
- Aleksandra Gostyńska
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Kamila Buzun
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland.
| | - Izabela Żółnowska
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Dorota Mańkowska-Wierzbicka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
| | - Anna Jelińska
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| |
Collapse
|
3
|
Feng C, Qiao C, Ji W, Pang H, Wang L, Feng Q, Ge Y, Rui M. In silico screening and in vivo experimental validation of 15-PGDH inhibitors from traditional Chinese medicine promoting liver regeneration. Int J Biol Macromol 2024; 274:133263. [PMID: 38901515 DOI: 10.1016/j.ijbiomac.2024.133263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/25/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH), which acts as a negative regulator of prostaglandin E2 (PGE2) levels and activity, represents a promising pharmacological target for promoting liver regeneration. In this study, we collected data on 15-PGDH homologous family proteins, their inhibitors, and traditional Chinese medicine (TCM) compounds. Leveraging machine learning and molecular docking techniques, we constructed a prediction model for virtual screening of 15-PGDH inhibitors from TCM compound library and successfully screened genistein as a potential 15-PGDH inhibitor. Through further validation, it was discovered that genistein considerably enhances liver regeneration by inhibiting 15-PGDH, resulting in a significant increase in the PGE2 level. Genistein's effectiveness suggests its potential as a novel therapeutic agent for liver diseases, highlighting this study's contribution to expanding the clinical applications of TCM.
Collapse
Affiliation(s)
- Chunlai Feng
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Chunxue Qiao
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Wei Ji
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Hui Pang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Li Wang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Qiuqi Feng
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Yingying Ge
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Mengjie Rui
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China.
| |
Collapse
|
4
|
Wang X, Hu X, Ye C, Zhao J, Tan SC, Zhou L, Zhao C, Wu KH, Yang X, Wei J, Yang M. Astragalus Polysaccharide Enhances Voriconazole Metabolism under Inflammatory Conditions through the Gut Microbiota. J Clin Transl Hepatol 2024; 12:481-495. [PMID: 38779521 PMCID: PMC11106349 DOI: 10.14218/jcth.2024.00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 05/25/2024] Open
Abstract
Background and Aims Voriconazole (VRC), a widely used antifungal drug, often causes hepatotoxicity, which presents a significant clinical challenge. Previous studies demonstrated that Astragalus polysaccharide (APS) can regulate VRC metabolism, thereby potentially mitigating its hepatotoxic effects. In this study, we aimed to explore the mechanism by which APS regulates VRC metabolism. Methods First, we assessed the association of abnormal VRC metabolism with hepatotoxicity using the Roussel Uclaf Causality Assessment Method scale. Second, we conducted a series of basic experiments to verify the promotive effect of APS on VRC metabolism. Various in vitro and in vivo assays, including cytokine profiling, immunohistochemistry, quantitative polymerase chain reaction, metabolite analysis, and drug concentration measurements, were performed using a lipopolysaccharide-induced rat inflammation model. Finally, experiments such as intestinal biodiversity analysis, intestinal clearance assessments, and Bifidobacterium bifidum replenishment were performed to examine the ability of B. bifidum to regulate the expression of the VRC-metabolizing enzyme CYP2C19 through the gut-liver axis. Results The results indicated that APS does not have a direct effect on hepatocytes. However, the assessment of gut microbiota function revealed that APS significantly increases the abundance of B. bifidum, which could lead to an anti-inflammatory response in the liver and indirectly enhance VRC metabolism. The dual-luciferase reporter gene assay revealed that APS can hinder the secretion of pro-inflammatory mediators and reduce the inhibitory effect on CYP2C19 transcription through the nuclear factor-κB signaling pathway. Conclusions The study offers valuable insights into the mechanism by which APS alleviates VRC-induced liver damage, highlighting its immunomodulatory influence on hepatic tissues and its indirect regulatory control of VRC-metabolizing enzymes within hepatocytes.
Collapse
Affiliation(s)
- Xiaokang Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, China
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Guangdong Medical University, Dongguan, Guangdong, China
- Department of Pharmacy, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Xianjing Hu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Guangdong Medical University, Dongguan, Guangdong, China
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Chunxiao Ye
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingqian Zhao
- Department of Pharmacy, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chenyu Zhao
- Department of Pharmacy, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Kit Hang Wu
- Department of Pharmacy, Nossa Senhora do Carmo-Lago Health Centre, Health Bureau, Macau, China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Jinbin Wei
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Maoxun Yang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, China
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, Guangdong Medical University, Dongguan, Guangdong, China
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
5
|
Shi Z, Guan N, Sun W, Sun T, Niu L, Li J, Ge J. Protective Effect of Levilactobacillus brevis Against Yersinia enterocolitica Infection in Mouse Model via Regulating MAPK and NF-κB Pathway. Probiotics Antimicrob Proteins 2022; 14:830-844. [PMID: 35665480 DOI: 10.1007/s12602-022-09957-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
Although the use of the probiotic bacterium Lactobacillus for the treatment and prevention of diseases caused by various pathogenic bacteria has received increasing attention in recent years, its mechanism remains incompletely understood. Levilactobacillus brevis 23017 is a select probiotic strain that can regulate the immunity of host animals and resist pathogen infections. In this study, we analyzed the effect of L. brevis 23017 on Yersinia enterocolitica intestinal infection in a BALB/c mouse model and discussed its underlying mechanism. We found that in the mouse model, L. brevis 23017 prevented the damage of villi in the small intestine and decelerated weight loss after Y. enterocolitica infection. Moreover, we focused on the mechanism of the protective effect of L. brevis 23017 from the perspective of the damage and repair of the intestinal mucosal barrier. We observed that L. brevis 23017 maintained a normal mucosal barrier by altering the expression of tight junction proteins. Notably, our results indicated that L. brevis 23017 effectively promoted the secretion of the intestine-specific secretory immunoglobulin A (SIgA) by B cells via regulating cytokines and oxidative damage levels. This mechanism may be the reason for its protective role in Y. enterocolitica infection. In addition, our results demonstrated that the mechanism of L. brevis 23017 was related to antibacterial colonization and inflammation regulation and closely related to antioxidative stress and SIgA promotion. The protective effect of L. brevis 23017 on mice was related to the signaling pathway protein p38 MAPK and the phosphorylation levels of NF-κB. Our study provided novel insight into the mechanism of Lactobacillus against pathogenic bacterial infections. Such insight is of great importance for the prevention, diagnosis, and treatment of related diseases.
Collapse
Affiliation(s)
- Ziqi Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Naiyu Guan
- Key Laboratory of Zoonoses Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Weijiao Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tianzhi Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Lingdi Niu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jinyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
- Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Harbin, 150030, China.
| |
Collapse
|
6
|
Paulino do Nascimento LC, Lacerda DC, Ferreira DJS, de Souza EL, de Brito Alves JL. Limosilactobacillus fermentum, Current Evidence on the Antioxidant Properties and Opportunities to be Exploited as a Probiotic Microorganism. Probiotics Antimicrob Proteins 2022; 14:960-979. [PMID: 35467236 DOI: 10.1007/s12602-022-09943-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2022] [Indexed: 12/14/2022]
Abstract
The unbalance in the production and removal of oxygen-reactive species in the human organism leads to oxidative stress, a physiological condition commonly linked to the occurrence of cancer, neurodegenerative, inflammatory, and metabolic disorders. The implications of oxidative stress in the gut have been associated with gut microbiota impairments and gut dysbiosis. Some lactobacilli strains have shown an efficient antioxidant system capable of protecting against oxidative stress and related-chronic diseases. Recently, in vitro and experimental studies and some clinical trials have demonstrated the efficacy of the administration of various Limosilactobacillus fermentum strains to modulate beneficially the host antioxidant system resulting in the amelioration of a variety of systemic diseases phenotypes. This review presents and discusses the currently available studies on identifying L. fermentum strains with anti-oxidant properties, their sources, range of the administered doses, and duration of the intervention in experiments with animals and clinical trials. This review strives to serve as a relevant and well-cataloged reference of L. fermentum strains with capabilities of inducing anti-oxidant effects and health-promoting benefits to the host, envisaging their broad applicability to disease control.
Collapse
Affiliation(s)
| | - Diego Cabral Lacerda
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - João Pessoa, Paraíba, Brazil
| | | | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - João Pessoa, Paraíba, Brazil
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I - João Pessoa, Paraíba, Brazil.
| |
Collapse
|
7
|
Xu S, Zhao M, Wang Q, Xu Z, Pan B, Xue Y, Dai Z, Wang S, Xue Z, Wang F, Xu C. Effectiveness of Probiotics and Prebiotics Against Acute Liver Injury: A Meta-Analysis. Front Med (Lausanne) 2021; 8:739337. [PMID: 34621765 PMCID: PMC8490661 DOI: 10.3389/fmed.2021.739337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Acute liver injury (ALI) is a clinical syndrome characterized by rapid loss of liver function, which may progress to life-threatening liver failure. We conducted this meta-analysis to examine the evidence on the effects of probiotics or prebiotics on ALI. Methods and Results: Several databases, including PubMed, EMBASE, and Cochrane Library, were scrutinized from the inception through February 2021 by combining key search terms, yielding 26 eligible studies, which concluded that modulation of gut microbiota significantly decreased aspartate transaminase [standardized mean difference (SMD): −1.51, 95% confidence interval (CI): −2.03 to −1.00], alanine aminotransferase (SMD: −1.42, 95% CI: −1.85 to −0.98), and bilirubin (SMD: −0.91, 95% CI: −1.33 to −0.49). In addition, administration of probiotics or prebiotics also promoted proliferation of Bifidobacterium (SMD: 1.21, 95% CI: −0.18 to 2.60) and inhibited Enterococcus (SMD: −1.00, 95% CI: −1.39 to −0.61), contributing to lower levels of endotoxin (SMD: −2.14, 95% CI: −2.91 to −1.37). Tight junction protein ZO-1 (SMD: 1.95, 95% CI: 0.14 to 3.76) was upregulated after intervention, thereby reducing bacterial translocation to the liver [odds ratio (OR) = 0.23, 95% CI: 0.13–0.44] and mesenteric lymph node (OR = 0.14, 95% CI: 0.08 to 0.26), with decreased tumor necrosis factor-α (SMD: −2.84, 95% CI: −3.76 to −1.93) and interleukin-6 (SMD: −2.62, 95% CI: −4.14 to −1.10). Oxidative stress was also relieved by reducing malondialdehyde (SMD: −1.83, 95% CI: −2.55 to −1.10) while elevating superoxide dismutase (SMD: 1.78, 95% CI: 1.00–2.55) and glutathione (SMD: 1.83, 95% CI: 0.76–2.91). Conclusion: Our findings suggest that probiotics and prebiotics could be a promising therapeutic strategy in ALI and possess a potential for clinical applications. Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=255888, CRD42021255888.
Collapse
Affiliation(s)
- Sheng Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Min Zhao
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinjian Wang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhihua Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binhui Pan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yilang Xue
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zebin Dai
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sisi Wang
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Zhanxiong Xue
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Changlong Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Cheng H, Huang H, Guo Z, Chang Y, Li Z. Role of prostaglandin E2 in tissue repair and regeneration. Am J Cancer Res 2021; 11:8836-8854. [PMID: 34522214 PMCID: PMC8419039 DOI: 10.7150/thno.63396] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Tissue regeneration following injury from disease or medical treatment still represents a challenge in regeneration medicine. Prostaglandin E2 (PGE2), which involves diverse physiological processes via E-type prostanoid (EP) receptor family, favors the regeneration of various organ systems following injury for its capabilities such as activation of endogenous stem cells, immune regulation, and angiogenesis. Understanding how PGE2 modulates tissue regeneration and then exploring how to elevate the regenerative efficiency of PGE2 will provide key insights into the tissue repair and regeneration processes by PGE2. In this review, we summarized the application of PGE2 to guide the regeneration of different tissues, including skin, heart, liver, kidney, intestine, bone, skeletal muscle, and hematopoietic stem cell regeneration. Moreover, we introduced PGE2-based therapeutic strategies to accelerate the recovery of impaired tissue or organs, including 15-hydroxyprostaglandin dehydrogenase (15-PGDH) inhibitors boosting endogenous PGE2 levels and biomaterial scaffolds to control PGE2 release.
Collapse
|
9
|
Spangler JR, Caruana JC, Medintz IL, Walper SA. Harnessing the potential of Lactobacillus species for therapeutic delivery at the lumenal-mucosal interface. Future Sci OA 2021; 7:FSO671. [PMID: 33815818 PMCID: PMC8015674 DOI: 10.2144/fsoa-2020-0153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lactobacillus species have been studied for over 30 years in their role as commensal organisms in the human gut. Recently there has been a surge of interest in their abilities to natively and recombinantly stimulate immune activities, and studies have identified strains and novel molecules that convey particular advantages for applications as both immune adjuvants and immunomodulators. In this review, we discuss the recent advances in Lactobacillus-related activity at the gut/microbiota interface, the efforts to probe the boundaries of the direct and indirect therapeutic potential of these bacteria, and highlight the continued interest in harnessing the native capacity for the production of biogenic compounds shown to influence nervous system activity. Taken together, these aspects underscore Lactobacillus species as versatile therapeutic delivery vehicles capable of effector production at the lumenal-mucosal interface, and further establish a foundation of efficacy upon which future engineered strains can expand.
Collapse
Affiliation(s)
- Joseph R Spangler
- National Research Council Postdoctoral Fellow sited in US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Julie C Caruana
- American Society for Engineering Education Postdoctoral Fellow sited in US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Igor L Medintz
- US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Scott A Walper
- US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| |
Collapse
|
10
|
Deng Q, Shi H, Luo Y, Zhao H, Liu N. Effect of dietary Lactobacilli mixture on Listeria monocytogenes infection and virulence property in broilers. Poult Sci 2020; 99:3655-3662. [PMID: 32616262 PMCID: PMC7597833 DOI: 10.1016/j.psj.2020.03.058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
The present study aimed to investigate the effect of probiotic Lactobacilli addition on Listeria monocytogenes load, inflammatory reaction, and virulence properties in broilers from 1 to 14 D of age. A total of 480 broiler chicks were randomly allocated to 4 treatments of 6 replicates each. All birds were infected with L. monocytogenes on the first day and supplemented an equal amount mixture of Lactobacillus acidophilus and Lactobacillus plantarum at doses of 0 (control), 106, 108, 1010 cfu/kg of diet. The results showed that on 7 and 14 D after administration, Lactobacilli addition at the 3 doses decreased (P < 0.05) L. monocytogenes loads in the cecum, skin, liver, and spleen by 0.065 to 0.933 log10 cfu, and the pathogen linearly reduced (P ≤ 0.015) with the increasing doses of probiotics in the skin. Serum cytokines including IL-1β, IL-6, tumor necrosis factor-α, and interferon-γ in probiotics treatments were decreased (P < 0.05) by 25.4 to 51.1%. Transcriptional levels of genes related to anti-inflammatory reactions including IL-10, hypoxia inducible factor 1 alpha (HIF1A), prostaglandin E receptor 2, and prostaglandin-endoperoxide synthase 2 in the intestinal mucosa were upregulated (P < 0.05) in Lactobacilli treatments, and linear and quadratic responses (P ≤ 0.019) were found on HIF1A. Furthermore, the probiotics attenuated (P < 0.05) listerial adhesion, pore-forming, and invasion properties by downregulating autolysin Ami, listeriolysin O, internalin A and B, and a linear (P = 0.006) dose response of probiotics was exhibited on flagellin. The findings indicate that dietary coadministration of L. acidophilus and L. plantarum can attenuate L. monocytogenes infection by depressing its intestinal inoculation, translocation, inflammatory reaction, and virulence property in broilers and suggest that the probiotics can be an alternative against listerial infection in broilers.
Collapse
Affiliation(s)
- Qingqing Deng
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471023, China
| | - Hanyi Shi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Yiran Luo
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471023, China
| | - Heping Zhao
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471023, China
| | - Ning Liu
- Department of Animal Production, Henan University of Science and Technology, Luoyang 471023, China; National Engineering Research Center of Biological Feed, Beijing 100008, China.
| |
Collapse
|
11
|
Cao C, Wu R, Zhu X, Li Y, Li M, An F, Wu J. Ameliorative effect of Lactobacillus plantarum WW-fermented soy extract on rat fatty liver via the PPAR signaling pathway. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
12
|
|
13
|
Jiang X, Gu S, Liu D, Zhao L, Xia S, He X, Chen H, Ge J. Lactobacillus brevis 23017 Relieves Mercury Toxicity in the Colon by Modulation of Oxidative Stress and Inflammation Through the Interplay of MAPK and NF-κB Signaling Cascades. Front Microbiol 2018; 9:2425. [PMID: 30369917 PMCID: PMC6194351 DOI: 10.3389/fmicb.2018.02425] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/21/2018] [Indexed: 02/06/2023] Open
Abstract
Aims: Lactobacillus strains have protective effects against heavy metals while relieving oxidative stress and modulating the immune response. Mechanisms that ameliorate heavy metal toxicity and the relationship between probiotics and gut barrier protection in the process of heavy metal pathogenesis was poorly understood. Methods and Results: In this study, Lactobacillus brevis 23017 (LAB, L. brevis 23017), a selected probiotics strain with strong mercury binding capacities, was applied to evaluate the efficiency against mercury toxicity in a mouse model. Histopathological results along with HE stains show that L. brevis 23017 protects the integrity of the small intestinal villus, which slows weight loss in response to Hg exposure. The qRT-PCR results demonstrate that L. brevis 23017 maintains a normal mucosal barrier via modulation of tight junction proteins. Importantly, the present study demonstrates that L. brevis 23017 effectively ameliorates injury of the small intestine by reducing intestinal inflammation and alleviating oxidative stress in animal models. Moreover, L. brevis 23017 blocks oxidative stress and inflammation through MAPK and NF-κB pathways, as shown by western blot. Conclusions: Together, these results reveal that L. brevis 23017 may have applications in the prevention and treatment of oral Hg exposure with fermented functional foods by protecting gut health in daily life.
Collapse
Affiliation(s)
- Xinpeng Jiang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Shanshan Gu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Di Liu
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, Harbin, China
| | - Lili Zhao
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin Veterinary Research Institute, Harbin, China
| | - Shuang Xia
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinmiao He
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, Harbin, China
| | - Hongyan Chen
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin Veterinary Research Institute, Harbin, China
| | - Junwei Ge
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
14
|
Uribe G, Villéger R, Bressollier P, Dillard RN, Worthley DL, Wang TC, Powell DW, Urdaci MC, Pinchuk IV. Lactobacillus rhamnosus GG increases cyclooxygenase-2 expression and prostaglandin E2 secretion in colonic myofibroblasts via a MyD88-dependent mechanism during homeostasis. Cell Microbiol 2018; 20:e12871. [PMID: 29920917 DOI: 10.1111/cmi.12871] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/12/2022]
Abstract
Prostaglandin E2 (PGE2 ) plays a critical role in intestinal mucosal tolerance and barrier integrity. Cyclooxygenase-2 (COX-2)-dependent PGE2 production involves mobilisation of arachidonic acid. Lactobacillus rhamnosus GG (LbGG) is one of the most widely used probiotics reported to colonise the colonic mucosa. LbGG contributes to the protection of the small intestine against radiation injury through the repositioning of mucosal COX-2 expressing cells. However, it is unknown if LbGG modulates PGE2 production in the colonic mucosa under homeostasis and the major cellular elements involved in these processes. Colonic epithelial and CD90+ mesenchymal stromal cells, also known as (myo) fibroblasts (CMFs), are abundant innate immune cells in normal colonic mucosa able to produce PGE2 . Herein, we tested the hypothesis that under colonic mucosal homeostasis, LbGG modulates the eicosanoid pathway resulting in increased PGE2 production in both epithelial and stromal cells. Among the five tested human colonic epithelial cell lines, only exposure of Caco-2 to LbGG for 24 hr led to the mobilisation of arachidonic acid with concomitant increase in the components within the leukotriene and COX-2-dependent PGE2 pathways. By contrast, CMFs isolated from the normal human colonic mucosa responded to LbGG with increased expression of COX-2 and PGE2 in the prostaglandin pathway, but not 5-LO in the leukotriene pathway. Oral gavage of C57BL/6 mice for 5 days with LbGG (5 × 108 Colony-Forming Unit (CFU)/dose) increased COX-2 expression in the colonic mucosa. The majority of cells upregulating COX-2 protein expression were located in the colonic lamina propria and colocalised with α-SMA+ cells corresponding to the CMF phenotype. This process was myeloid differentiation factor-88-dependent, because silencing of myeloid differentiation factor-88 expression in CMFs abrogated LbGG-induced upregulation of COX-2 in culture and in vivo. Taken together, our data suggest that LbGG increases release of COX-2-mediated PGE2 , contributing to the maintenance of mucosal homeostasis in the colon and CMFs are among the major contributors to this process.
Collapse
Affiliation(s)
- Gabriela Uribe
- Departments of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Romain Villéger
- Departments of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Philippe Bressollier
- Laboratoire de Microbiologie, Bordeaux Sciences Agro, University of Bordeaux, Gradignan, France
| | - Rachel N Dillard
- Departments of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Daniel L Worthley
- Cancer Theme, University of Adelaide and SAHMRI, Adelaide, Australia
| | - Timothy C Wang
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Don W Powell
- Departments of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Maria C Urdaci
- Laboratoire de Microbiologie, Bordeaux Sciences Agro, University of Bordeaux, Gradignan, France
| | - Irina V Pinchuk
- Departments of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA.,Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
15
|
Bluemel S, Williams B, Knight R, Schnabl B. Precision medicine in alcoholic and nonalcoholic fatty liver disease via modulating the gut microbiota. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1018-G1036. [PMID: 27686615 PMCID: PMC5206291 DOI: 10.1152/ajpgi.00245.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/25/2016] [Indexed: 02/08/2023]
Abstract
Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) represent a major health burden in industrialized countries. Although alcohol abuse and nutrition play a central role in disease pathogenesis, preclinical models support a contribution of the gut microbiota to ALD and NAFLD. This review describes changes in the intestinal microbiota compositions related to ALD and NAFLD. Findings from in vitro, animal, and human studies are used to explain how intestinal pathology contributes to disease progression. This review summarizes the effects of untargeted microbiome modifications using antibiotics and probiotics on liver disease in animals and humans. While both affect humoral inflammation, regression of advanced liver disease or mortality has not been demonstrated. This review further describes products secreted by Lactobacillus- and microbiota-derived metabolites, such as fatty acids and antioxidants, that could be used for precision medicine in the treatment of liver disease. A better understanding of host-microbial interactions is allowing discovery of novel therapeutic targets in the gut microbiota, enabling new treatment options that restore the intestinal ecosystem precisely and influence liver disease. The modulation options of the gut microbiota and precision medicine employing the gut microbiota presented in this review have excellent prospects to improve treatment of liver disease.
Collapse
Affiliation(s)
- Sena Bluemel
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Brandon Williams
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Rob Knight
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, La Jolla, California; and
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California;
- Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|