1
|
Chen L, Wang X, Wang S, Liu W, Song Z, Liao H. The impact of gut microbiota on the occurrence, treatment, and prognosis of ischemic stroke. Neurobiol Dis 2025; 207:106836. [PMID: 39952411 DOI: 10.1016/j.nbd.2025.106836] [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: 12/30/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025] Open
Abstract
Ischemic stroke (IS) is a cerebrovascular disease that predominantly affects middle-aged and elderly populations, exhibiting high mortality and disability rates. At present, the incidence of IS is increasing annually, with a notable trend towards younger affected individuals. Recent discoveries concerning the "gut-brain axis" have established a connection between the gut and the brain. Numerous studies have revealed that intestinal microbes play a crucial role in the onset, progression, and outcomes of IS. They are involved in the entire pathophysiological process of IS through mechanisms such as chronic inflammation, neural regulation, and metabolic processes. Although numerous studies have explored the relationship between IS and intestinal microbiota, comprehensive analyses of specific microbiota is relatively scarce. Therefore, this paper provides an overview of the typical changes in gut microbiota following IS and investigates the role of specific microorganisms in this context. Additionally, it presents a comprehensive analysis of post-stroke microbiological therapy and the relationship between IS and diet. The aim is to identify potential microbial targets for therapeutic intervention, as well as to highlight the benefits of microbiological therapies and the significance of dietary management. Overall, this paper seeks to provide key strategies for the treatment and management of IS, advocating for healthy diets and health programs for individuals. Meanwhile, it may offer a new perspective on the future interdisciplinary development of neurology, microbiology and nutrition.
Collapse
Affiliation(s)
- Liying Chen
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Shiqi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Weili Liu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Huiling Liao
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
2
|
Wang S, Nie Z, Zhu L, Wu Y, Wen Y, Deng F, Zhao L. Probiotic Characteristics and the Anti-Inflammatory Effects of Lactiplantibacillus plantarum Z22 Isolated from Naturally Fermented Vegetables. Microorganisms 2024; 12:2159. [PMID: 39597548 PMCID: PMC11596721 DOI: 10.3390/microorganisms12112159] [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: 09/11/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Currently, there is increasing interest in the commercial utilization of probiotics isolated from traditional fermented food products. Therefore, this study aimed to investigate the probiotic potential of Lactiplantibacillus plantarum (L. plantarum) Z22 isolated from naturally fermented mustard. The results suggest that L. plantarum Z22 exhibits good adhesion ability, antibacterial activity, safety, and tolerance to acidic conditions and bile salts. We further determined the anti-inflammatory mechanism and properties of L. plantarum Z22 and found that L. plantarum Z22 could significantly reduce the secretion of pro-inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and the expression of the pro-inflammatory mediator cyclooxygenase-2 (COX-2) protein in LPS-induced RAW 264.7 cells. In addition, L. plantarum Z22 also effectively inhibited the signaling pathways of nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPKs). This effect can be attributed to a decrease in the levels of reactive oxygen species (ROS) and increased heme oxygenase-1 (HO-1) expression. Moreover, whole-genome sequencing revealed that L. plantarum Z22 contains gene-encoding proteins with anti-inflammatory functions, such as beta-glucosidase (BGL) and pyruvate kinase (PK), as well as antioxidant functions, including thioredoxin reductase (TrxR), tyrosine-protein phosphatase, and ATP-dependent intracellular proteases ClpP. In summary, these results indicated that L. plantarum Z22 can serve as a potential candidate probiotic for use in fermented foods such as yogurt (starter cultures), providing a promising strategy for the development of functional foods to prevent chronic diseases.
Collapse
Affiliation(s)
- Shiyu Wang
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Ziyu Nie
- College of Animal Science and Technology, Hunan Biological Electromechanical Vocational College, Changsha 410128, China;
| | - Li Zhu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Yanyang Wu
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Yashi Wen
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Fangming Deng
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| | - Lingyan Zhao
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China; (S.W.); (L.Z.); (Y.W.); (Y.W.)
| |
Collapse
|
3
|
Qin M, Xing Y, Sun M, Ma L, Li X, Ma F, Li D, Duan C. An Exploration of the Antioxidative and Anti-Inflammatory Role of Lactiplantibacillus plantarum 106 via Improving Mitochondrial Function. Foods 2024; 13:1981. [PMID: 38998487 PMCID: PMC11241742 DOI: 10.3390/foods13131981] [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: 05/21/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
In this present study, bioinformatics analysis and the experimental validation method were used to systematically explore the antioxidant activity and anti-inflammatory effect of Lactiplantibacillus plantarum A106, which was isolated from traditional Chinese pickles, on lipopolysaccharide (LPS)-induced RAW264.7 macrophages. L. plantarum A106 had a good scavenging ability for DPPH, ABTS, and hydroxyl radicals. Furthermore, L. plantarum A106 could increase the activity of RAW264.7 macrophages; raise the SOD and GSH levels, with or without LPS sensitization; or decrease the MDA, TNF-α, and IL-6 levels. In order to deeply seek the antioxidant and anti-inflammatory role and mechanism, bioinformatic analysis, including GO, KEGG, and GSEA analysis, was used to conduct an in-depth analysis, and the results showed that the LPS treatment of RAW264.7 macrophages significantly upregulated inflammatory-related genes and revealed an enrichment in the inflammatory signaling pathways. Additionally, a network analysis via the Cytoscape software (version 3.9.1) identified key central genes and found that LPS also disturbed apoptosis and mitochondrial function. Based on the above bioinformatics analysis, the effects of L. plantarum A106 on inflammation-related gene expression, mitochondrial function, apoptosis, etc., were detected. The results indicated that L. plantarum A106 restored the declined expression levels of crucial genes like TNF-α and IL-6; mitochondrial membrane potential; and apoptosis and the expression of apoptosis-related genes, Bcl-2, Caspase-3, and Bax. These results suggest that L. plantarum A106 exerts antioxidant activity and anti-inflammatory effects through regulating inflammatory and apoptosis-related gene expression, restoring the mitochondrial membrane potential.
Collapse
Affiliation(s)
- Mengchun Qin
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Yinfei Xing
- College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Maocheng Sun
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Lin Ma
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Xiaolei Li
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Fumin Ma
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Dan Li
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| | - Cuicui Duan
- Key Laboratory of Agro-Products Processing Technology, Education Department of Jilin Province, Changchun University, 6543 Weixing Road, Changchun 130022, China
| |
Collapse
|
4
|
Li J, Liu H, Fu H, Yang Y, Wu Z. An Isofibrous Diet with Fiber Konjac Glucomannan Ameliorates Salmonella typhimurium-Induced Colonic Injury by Regulating TLR2-NF-κB Signaling and Intestinal Microbiota in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13415-13430. [PMID: 38824655 DOI: 10.1021/acs.jafc.4c03019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study aimed to investigate the hypothesis that dietary konjac glucomannan (KGM) could alleviate Salmonella typhimurium-induced colitis by modulating intestinal microbiota. Mice were fed an isocaloric and isofibrous diet supplemented with either 7% KGM or cellulose and were treated with 5 × 108 CFU of S. typhimurium. The results showed that KGM had an average molecular weight of 936 kDa and predominantly consisted of mannose and glucose at a molar ratio of 1:1.22. In vivo studies demonstrated that dietary KGM effectively mitigated colonic lesions, oxidative stress, disruption of tight junction protein 2 and occludin, and the inflammatory response induced by S. typhimurium. Moreover, KGM administration alleviated the dramatic upregulation of toll-like receptor 2 (TLR2) and phosphonuclear factor κB (NF-κB) protein abundance, induced by Salmonella treatment. Notably, dietary KGM restored the reduced Muribaculaceae and Lactobacillus abundance and increased the abundance of Blautia and Salmonella in S. typhimurium-infected mice. Spearman correlation analysis revealed that the gut microbiota improved by KGM contribute to inhibit inflammation and oxidative stress. These results demonstrated the protective effects of dietary KGM against colitis by modulating the gut microbiota and the TLR2-NF-κB signaling pathway in response to Salmonella infection.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, P. R. China
| | - Haozhen Liu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, P. R. China
| | - Huiyang Fu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, P. R. China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, P. R. China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
5
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
6
|
Wong WY, Chan BD, Cho PT, Leung TW, Tai WCS. Beneficial and immunomodulatory effects of heat-killed Lactobacillus plantarum L137 in normal and acute colitis mice. J Funct Foods 2024; 116:106167. [DOI: 10.1016/j.jff.2024.106167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
7
|
Wang J, Zhang H, Yuan H, Chen S, Yu Y, Zhang X, Gao Z, Du H, Li W, Wang Y, Xia P, Wang J, Song M. Prophylactic Supplementation with Lactobacillus Reuteri or Its Metabolite GABA Protects Against Acute Ischemic Cardiac Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307233. [PMID: 38487926 PMCID: PMC11095141 DOI: 10.1002/advs.202307233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/06/2023] [Indexed: 05/16/2024]
Abstract
The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well-studied probiotic that can colonize mammals including humans with known cholesterol-lowering properties and anti-inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ-aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS-stimulated bone marrow-derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage-mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.
Collapse
Affiliation(s)
- Jiawan Wang
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- Beijing Chao‐Yang HospitalDepartment of AnesthesiologyBeijing100020China
| | - Hao Zhang
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hailong Yuan
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifeng475004China
| | - Siqi Chen
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Ying Yu
- University of Chinese Academy of SciencesBeijing100049China
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyChinese Academy of SciencesBeijing100101China
| | - Xuan Zhang
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyChinese Academy of SciencesBeijing100101China
| | - Zeyu Gao
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
| | - Heng Du
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Weitao Li
- Department of ImmunologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifeng475004China
| | - Pengyan Xia
- Department of ImmunologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Jun Wang
- University of Chinese Academy of SciencesBeijing100049China
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyChinese Academy of SciencesBeijing100101China
| | - Moshi Song
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
8
|
Zhou K, Xie J, Su Y, Fang J. Lactobacillus reuteri for chronic periodontitis: focus on underlying mechanisms and future perspectives. Biotechnol Genet Eng Rev 2024; 40:381-408. [PMID: 36856460 DOI: 10.1080/02648725.2023.2183617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023]
Abstract
Chronic periodontitis is a common oral disorder caused by pathogenic bacteria. Despite the wide use of antibiotics as the conventional adjunctive treatment, the challenges of increased antibiotic resistance and limited therapeutic effect receive considerable attention and the developments of alternative treatments gain increasing consideration. Growing evidence showed that Lactobacillus reuteri (LR) may represent a promising alternative adjunct for chronic periodontitis. It can attenuate inflammation and reduce tissue disruption. LR-assisted treatment has been shown to be effective and relatively safe in multiple clinical trials, and accumulating evidence suggests its significant biological roles. In the current review, we focus on capturing the underlying mechanisms of LR involved in chronic periodontitis, thereby representing a scientific foundation for LR-assisted therapy. Furthermore, we point out the challenges and future directions for further clinical trials to improve the clinical applicability for LR.
Collapse
Affiliation(s)
- Keyi Zhou
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| | - Jiaman Xie
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| | - Yuan Su
- Department of Periodontology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| | - Jingxian Fang
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
9
|
Jiang S, Si J, Mo J, Zhang S, Chen K, Gao J, Xu D, Bai L, Lan G, Liang J. Integrated Microbiome and Serum Metabolome Analysis Reveals Molecular Regulatory Mechanisms of the Average Daily Weight Gain of Yorkshire Pigs. Animals (Basel) 2024; 14:278. [PMID: 38254447 PMCID: PMC10812420 DOI: 10.3390/ani14020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The average daily weight gain (ADG) is considered a crucial indicator for assessing growth rates in the swine industry. Therefore, investigating the gastrointestinal microbiota and serum metabolites influencing the ADG in pigs is pivotal for swine breed selection. This study involved the inclusion of 350 purebred Yorkshire pigs (age: 90 ± 2 days; body weight: 41.20 ± 4.60 kg). Concurrently, serum and fecal samples were collected during initial measurements of blood and serum indices. The pigs were categorized based on their ADG, with 27 male pigs divided into high-ADG (HADG) and low-ADG (LADG) groups based on their phenotype values. There were 12 pigs in LADG and 15 pigs in HADG. Feces and serum samples were collected on the 90th day. Microbiome and non-targeted metabolomics analyses were conducted using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC-MS). Pearson correlation, with Benjamini-Hochberg (BH) adjustment, was employed to assess the associations between these variables. The abundance of Lactobacillus and Prevotella in LADG was significantly higher than in HADG, while Erysipelothrix, Streptomyces, Dubosiella, Parolsenella, and Adlercreutzia in LADG were significantly lower than in HADG. The concentration of glutamine, etiocholanolone glucuronide, and retinoyl beta-glucuronide in LADG was significantly higher than in HADG, while arachidonic acid, allocholic acid, oleic acid, phenylalanine, and methyltestosterone in LADG were significantly lower than in HADG. The Lactobacillus-Streptomyces networks (Lactobacillus, Streptomyces, methyltestosterone, phenylalanine, oleic acid, arachidonic acid, glutamine, 3-ketosphingosine, L-octanoylcarnitine, camylofin, 4-guanidinobutyrate 3-methylcyclopentadecanone) were identified as the most influential at regulating swine weight gain. These findings suggest that the gastrointestinal tract regulates the daily weight gain of pigs through the network of Lactobacillus and Streptomyces. However, this study was limited to fecal and serum samples from growing and fattening boars. A comprehensive consideration of factors affecting the daily weight gain in pig production, including gender, parity, season, and breed, is warranted.
Collapse
Affiliation(s)
- Shan Jiang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Jinglei Si
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
- Guangxi State Farms Yongxin Animal Husbandary Group Co., Ltd., Nanning 530022, China
| | - Jiayuan Mo
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Shuai Zhang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Kuirong Chen
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Jiuyu Gao
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Di Xu
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Lijing Bai
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Ganqiu Lan
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| | - Jing Liang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China; (S.J.); (J.S.); (J.M.); (S.Z.); (K.C.); (J.G.); (D.X.); (G.L.)
| |
Collapse
|
10
|
Petrariu OA, Barbu IC, Niculescu AG, Constantin M, Grigore GA, Cristian RE, Mihaescu G, Vrancianu CO. Role of probiotics in managing various human diseases, from oral pathology to cancer and gastrointestinal diseases. Front Microbiol 2024; 14:1296447. [PMID: 38249451 PMCID: PMC10797027 DOI: 10.3389/fmicb.2023.1296447] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
The imbalance of microbial composition and diversity in favor of pathogenic microorganisms combined with a loss of beneficial gut microbiota taxa results from factors such as age, diet, antimicrobial administration for different infections, other underlying medical conditions, etc. Probiotics are known for their capacity to improve health by stimulating the indigenous gut microbiota, enhancing host immunity resistance to infection, helping digestion, and carrying out various other functions. Concurrently, the metabolites produced by these microorganisms, termed postbiotics, which include compounds like bacteriocins, lactic acid, and hydrogen peroxide, contribute to inhibiting a wide range of pathogenic bacteria. This review presents an update on using probiotics in managing and treating various human diseases, including complications that may emerge during or after a COVID-19 infection.
Collapse
Affiliation(s)
- Oana-Alina Petrariu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Ilda Czobor Barbu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Georgiana Alexandra Grigore
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Roxana-Elena Cristian
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Grigore Mihaescu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|
11
|
Zhang Y, Hu J, Song X, Dai J, Tang Z, Huang G, Jiao W, Wu Y, Wang C, Du L, Jin Y. The effects of Lactobacillus reuteri microcapsules on radiation-induced brain injury by regulating the gut microenvironment. Food Funct 2023; 14:10041-10051. [PMID: 37843434 DOI: 10.1039/d3fo03008c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
A radiation-induced brain injury (RIBI) is a major adverse event following radiotherapy of malignant tumors. RIBI would affect cognitive function, leading to a series of complications and even death. However, the pathogenesis of RIBI is still unclear, and it still lacks specific therapeutic drugs. The gut-brain bidirectional communication may be mediated by various microbiota and metabolites in the gastrointestinal tract. Probiotics are closely related to physiological health. The theory of the gut-brain axis provides us with a new idea to improve the gut microenvironment by supplementing probiotics against RIBI. Here, Lactobacillus reuteri microcapsules (LMCs) were prepared, which were predominantly irregular spheres with a rough surface under a scanning electron microscope and a narrow size distribution ranging from 20 to 700 μm. The transmission electron microscopy images showed that the structure of microcapsules containing Lactobacillus reuteri (L. reuteri) was a core and shell structure. The survival of L. reuteri in microcapsules was significantly more than that of free L. reuteri in the simulated stomach environment of pH 1.2. 16S rDNA sequencing showed that LMCs observably increased the relative abundance of Lactobacillus in RIBI mice. More importantly, compared with the RIBI model mice, the behavior of RIBI mice treated with LMCs was significantly improved. In addition, LMCs greatly alleviated the pathological damage of the hippocampus and intestines in the mice after irradiation and reduced the level of TNF-α and IL-6 in vivo. Generally, LMCs are a promising oral preparation, which provide new ideas and methods for the treatment of RIBI.
Collapse
Affiliation(s)
- Yizhi Zhang
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jinglu Hu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
| | - Xingshuang Song
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing Dai
- Information Department, General Hospital of Western Zone, Chengdu 610083, China
| | - Ziyan Tang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Guiyu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
| | - Wencheng Jiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Hebei University, Baoding 071000, China
| | - Yanping Wu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chenyun Wang
- The Fourth Clinical Center Affiliated to Chinese PLA General Hospital, Beijing 100048, China
| | - Lina Du
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
- Hebei University, Baoding 071000, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Pharmaceutical College of Henan University, Kaifeng 475004, China
| |
Collapse
|
12
|
Shen J, Wang S, Huang Y, Wu Z, Han S, Xia H, Chen H, Li L. Lactobacillus reuteri Ameliorates Lipopolysaccharide-Induced Acute Lung Injury by Modulating the Gut Microbiota in Mice. Nutrients 2023; 15:4256. [PMID: 37836540 PMCID: PMC10574429 DOI: 10.3390/nu15194256] [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: 09/14/2023] [Revised: 09/25/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Acute lung injury (ALI) causes lung inflammation and edema as well as resulting in gut microbiota disorder. Probiotics, however, can improve the gut microbiota composition and modulate its immune response, playing an important role in ALI pathogenesis. Therefore, our study aims to investigate the effect of Lactobacillus reuteri on Lipopolysaccharide (LPS)-induced ALI in mice and to probe the mechanism of its synergistic modulatory effect on the lungs and intestines. We assessed the therapeutic effects of L. reuteri in the ALI mouse model by histopathology, alveolar lavage fluid and serum inflammatory factor analysis and explored microbiome and transcriptome alterations. L. reuteri intervention effectively attenuated lung tissue injury and significantly reduced the LPS-induced inflammatory response and macrophage and neutrophil infiltration. Additionally, L. reuteri improved the intestinal barrier function and remodeled the disordered microbiota. In conclusion, our study showed that L. reuteri attenuated the inflammatory response, ameliorated the pulmonary edema, repaired the intestinal barrier, and remodeled the gut microbiota in ALI mice. This study provides new perspectives on the clinical treatment of ALI.
Collapse
Affiliation(s)
- Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Shuting Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yong Huang
- Department of Infectious Disease, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310022, China
| | - Zhengjie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Hui Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| |
Collapse
|
13
|
El Far MS, Zakaria AS, Kassem MA, Wedn A, Guimei M, Edward EA. Promising biotherapeutic prospects of different probiotics and their derived postbiotic metabolites: in-vitro and histopathological investigation. BMC Microbiol 2023; 23:122. [PMID: 37138240 PMCID: PMC10155454 DOI: 10.1186/s12866-023-02866-1] [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: 02/21/2023] [Accepted: 04/19/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Probiotics and their derived postbiotics, as cell-free supernatants (CFS), are gaining a solid reputation owing to their prodigious health-promoting effects. Probiotics play a valuable role in the alleviation of various diseases among which are infectious diseases and inflammatory disorders. In this study, three probiotic strains, Lactiplantibacillus plantarum, Lacticaseibacillus rhamnosus, and Pediococcus acidilactici, were isolated from marketed dietary supplements. The antimicrobial activity of the isolated probiotic strains as well as their CFS was investigated. The neutralized CFS of the isolated probiotics were tested for their antibiofilm potential. The anti-inflammatory activity of the isolated Lactobacillus spp., together with their CFS, was studied in the carrageenan-induced rat paw edema model in male Wistar rats. To the best of our knowledge, such a model was not previously experimented to evaluate the anti-inflammatory activity of the CFS of probiotics. The histopathological investigation was implemented to assess the anti-inflammatory prospect of the isolated L. plantarum and L. rhamnosus strains as well as their CFS. RESULTS The whole viable probiotics and their CFS showed variable growth inhibition of the tested indicator strains using the agar overlay method and the microtiter plate assay, respectively. When tested for virulence factors, the probiotic strains were non-hemolytic lacking both deoxyribonuclease and gelatinase enzymes. However, five antibiotic resistance genes, blaZ, ermB, aac(6')- aph(2"), aph(3'')-III, and vanX, were detected in all isolates. The neutralized CFS of the isolated probiotics exhibited an antibiofilm effect as assessed by the crystal violet assay. This effect was manifested by hindering the biofilm formation of the tested Staphylococcus aureus and Pseudomonas aeruginosa clinical isolates in addition to P. aeruginosa PAO1 strain. Generally, the cell cultures of the two tested probiotics moderately suppressed the acute inflammation induced by carrageenan compared to indomethacin. Additionally, the studied CFS relatively reduced the inflammatory changes compared to the inflammation control group but less than that observed in the case of the probiotic cultures treated groups. CONCLUSIONS The tested probiotics, along with their CFS, showed promising antimicrobial and anti-inflammatory activities. Thus, their safety and their potential use as biotherapeutics for bacterial infections and inflammatory conditions are worthy of further investigation.
Collapse
Affiliation(s)
- Mona S El Far
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Azza S Zakaria
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mervat A Kassem
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Abdalla Wedn
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maha Guimei
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eva A Edward
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
14
|
Li Y, Li P, Yu X, Zheng X, Gu Q. Exploitation of In Vivo-Emulated In Vitro System in Advanced Food Science. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37023249 DOI: 10.1021/acs.jafc.2c07289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Reasonable model construction contributes to the accuracy of experimental results. Multiple in vivo models offer reliable choices for effective evaluation, whereas their applications are hampered due to adverse features including high time-consumption, high cost and ethical contradictions. In vivo-emulated in vitro systems (IVE systems) have experienced rapid development and have been brought into food science for about two decades. IVE systems' flexibly gathers the strengths of in vitro and in vivo models into one, reflecting the results in an efficient, systematic and interacted manner. In this review, we comprehensively reviewed the current research progress of IVE systems based on the literature published in the recent two decades. By categorizing the IVE systems into 2D coculture models, spheroids and organoids, their applications were systematically summarized and typically exemplified. The pros and cons of IVE systems were also thoroughly discussed, drawing attention to present challenges and inspiring potential orientation and future perspectives. The wide applicability and multiple possibilities suggest IVE systems as an effective and persuasive platform in the future of advanced food science.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, 310018, Zhejiang, People's Republic of China
| | - Ping Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, 310018, Zhejiang, People's Republic of China
| | - Xin Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, People's Republic of China
- Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, and National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, People's Republic of China
- Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, and National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, 310018, Zhejiang, People's Republic of China
| |
Collapse
|
15
|
Multi-Species Probiotic Strain Mixture Enhances Intestinal Barrier Function by Regulating Inflammation and Tight Junctions in Lipopolysaccharides Stimulated Caco-2 Cells. Microorganisms 2023; 11:microorganisms11030656. [PMID: 36985228 PMCID: PMC10056128 DOI: 10.3390/microorganisms11030656] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Although leaky gut syndrome is not recognized as an official diagnosis for human diseases, it is now believed that dysfunction of the cell barrier causes increased permeability of intestinal epithelial cells leading to this condition. Probiotics have been widely used to improve gut health, and studies have investigated the relevance of protecting the intestinal barrier by taking probiotic strains in vitro and in vivo. However, most studies have restricted the use of single or several probiotic strains and do not consider commercially available probiotic products composed of multi-species. In this study, we provide experimental evidence that a multi-species probiotic mixture composed of eight different strains and a heat-treated probiotic strain is effective in preventing leaky gut conditions. We employed an in vitro co-culture model system utilizing two different differentiated cell lines to mimic human intestinal tissue. The integrity of epithelial barrier function was protected by the preserving the occludin protein level and activating the AMPK signaling pathway, associated with tight junctions (TJs), through treatment with the probiotic strain mixture in Caco-2 cells. Moreover, we confirmed that application of the multi-species probiotic mixture reduced the expression of proinflammatory cytokine genes by inhibiting NFκB signaling pathway when artificial inflammation was induced in an in vitro co-culture model system. Finally, we proved that the epithelial permeability measured by trans-epithelial electrical resistance (TEER) was significantly decreased in the probiotic mixture treated cells, indicating that the integrity of the epithelial barrier function was not compromised. The multi-species probiotic strain mixture exhibited the protective effect on the integrity of intestinal barrier function via enhancing TJ complexes and reducing inflammatory responses in the human intestinal cells.
Collapse
|
16
|
Sheldon JM, Alonso N. The Therapeutic Benefits of Single and Multi-Strain Probiotics on Mean Daily Crying Time and Key Inflammatory Markers in Infantile Colic. Cureus 2022; 14:e28363. [PMID: 36168359 PMCID: PMC9506670 DOI: 10.7759/cureus.28363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Infantile colic is a functional gastrointestinal disorder in which a healthy infant displays paroxysms of intense crying or fussiness. Although this condition is self-limited, it causes significant distress for parents and may be linked to long-term health concerns for children. The microbiome of infants with colic has been correlated with increased dysbiosis or imbalance of commensal bacteria. This dysbiosis may ultimately lead to changes in infants’ immunological profiles, favoring markers linked to inflammation, including specific cytokines, calprotectin, and genetic markers. Therapeutic regimens such as probiotics may be helpful in modifying the gut microbial composition, thereby influencing the presence of inflammatory markers and potentially reducing colic symptoms in infants. This review provides a summary of the findings from 10 randomized, placebo-controlled, double-blinded studies conducted in the past five years with the aim of examining the potential therapeutic benefits of probiotics in infantile colic. The articles were selected through PubMed and Google Scholar using the keywords infantile colic, microbiome, probiotics, cytokines, dysbiosis, inflammatory markers, and lactobacilli. We summarize the results of these studies to explore the potential anti-inflammatory therapeutic benefits of single and multi-strain probiotic formulations on daily crying time and key inflammatory markers in infants with colic. The research largely shows the beneficial role of probiotics, largely of the lactobacillus genus, in the reduction of colic symptoms and the reduction of key inflammatory markers. However, some studies demonstrated an insignificant effect of certain probiotic strains in symptom management. Further research is necessary to better understand the anti-inflammatory properties of probiotics and determine the role this could have on the manifestation of colic in infants.
Collapse
|
17
|
Effects of the Probiotic, Lactobacillus delbrueckii subsp. bulgaricus, as a Substitute for Antibiotics on the Gastrointestinal Tract Microbiota and Metabolomics Profile of Female Growing-Finishing Pigs. Animals (Basel) 2022; 12:ani12141778. [PMID: 35883325 PMCID: PMC9311557 DOI: 10.3390/ani12141778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Lactobacillus delbrueckii subsp. bulgaricus (LDB) is an approved feed additive on the Chinese ‘Approved Feed Additives’ list. However, the possibility of LDB as an antibiotic replacement remains unclear. Particularly, the effect of LDB on microbiota and metabolites in the gastrointestinal tract (GIT) requires further explanation. This study aimed to identify the microbiota and metabolites present in fecal samples and investigate the relationship between the microbiota and metabolites to evaluate the potential of LDB as an antibiotic replacement in pig production. A total of 42 female growing-finishing pigs were randomly allocated into the antibiotic group (basal diet + 75 mg/kg aureomycin) and LDB (basal diet + 3.0 × 109 cfu/kg LDB) groups. Fecal samples were collected on days 0 and 30. Growth performance was recorded and assessed. 16S rRNA sequencing and liquid chromatography-mass spectrometry-based non-targeted metabolomics approaches were used to analyze the differences in microbiota and metabolites. Associations between the differences were calculated using Spearman correlations with the Benjamini−Hochberg adjustment. The LDB diet had no adverse effect on feed efficiency but slightly enhanced the average daily weight gain and average daily feed intake (p > 0.05). The diet supplemented with LDB increased Lactobacillus abundance and decreased that of Prevotellaceae_NK3B31_group spp. Dietary-supplemented LDB enhanced the concentrations of pyridoxine, tyramine, D-(+)-pyroglutamic acid, hypoxanthine, putrescine and 5-hydroxyindole-3-acetic acid and decreased the lithocholic acid concentration. The Lactobacillus networks (Lactobacillus, Peptococcus, Ruminococcaceae_UCG-004, Escherichia-Shigella, acetophenone, tyramine, putrescine, N-methylisopelletierine, N1-acetylspermine) and Prevotellaceae_NK3B31_group networks (Prevotellaceae_NK3B31_group, Treponema_2, monolaurin, penciclovir, N-(5-acetamidopentyl)acetamide, glycerol 3-phosphate) were the most important in the LDB effect on pig GIT health in our study. These findings indicate that LDB may regulate GIT function through the Lactobacillus and Prevotellaceae_NK3B31_group networks. However, our results were restrained to fecal samples of female growing-finishing pigs; gender, growth stages, breeds and other factors should be considered to comprehensively assess LDB as an antibiotic replacement in pig production.
Collapse
|
18
|
Zhang Y, Qin S, Song Y, Yuan J, Hu S, Chen M, Li L. Alginate Oligosaccharide Alleviated Cisplatin-Induced Kidney Oxidative Stress via Lactobacillus Genus-FAHFAs-Nrf2 Axis in Mice. Front Immunol 2022; 13:857242. [PMID: 35432359 PMCID: PMC9010505 DOI: 10.3389/fimmu.2022.857242] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/09/2022] [Indexed: 11/19/2022] Open
Abstract
Alginate oligosaccharide is the depolymerized product of alginate, a natural extract of brown algae, which is associated with beneficial health effects. Here, we aimed to investigate the mechanism via which alginate oligosaccharides improve kidney oxidative damage and liver inflammation induced by cisplatin chemotherapy via the gut microbiota. C57BL/6J mice were treated with cisplatin were administered alginate oligosaccharide via gavage for 3 weeks. Compared to that observed in the cisplatin chemotherapy group without intragastric administration of alginate oligosaccharide, liver inflammation improved in the alginate oligosaccharide group, indicated by reduction in lipopolysaccharide and interleukin-1β (IL-1β) levels. This was accompanied by improvement in the oxidative stress of mice kidneys, indicated by the increase in the levels of superoxide dismutase (SOD), catalase (CAT) and nuclear NF-E2-related factor 2 (Nrf2) in renal tissue, and reduction in the levels of malondialdehyde (MDA) in renal tissue and serum creatinine (Cr) to the levels of the normal control group. Alginate oligosaccharide intervention increased the concentration of fatty acid esters of hydroxy fatty acids (FAHFAs). Alginate oligosaccharide regulated the composition of the intestinal microbial community and promoted Lactobacillus stains, such as Lactobacillus johnsonii and Lactobacillus reuteri. Spearman analysis showed that 5 members of FAHFAs concentrations were positively correlated with Lactobacillus johnsonii and Lactobacillus reuteri abundance. We observed that alginate oligosaccharide increased FAHFAs producing-related bacterial abundance and FAHFAs levels, enhanced the levels of SOD and CAT in kidney tissue, and reduced the levels of MDA via activating Nrf2, thereby ameliorating the renal redox injury caused by cisplatin chemotherapy.
Collapse
Affiliation(s)
- Yubing Zhang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China
- College of Life Sciences, Yantai University, Yantai, China
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Yipeng Song
- Department of Radiotherapy, Yantai Yuhuangding Hospital, Yantai, China
| | - Jingyi Yuan
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Shanliang Hu
- Department of Radiotherapy, Yantai Yuhuangding Hospital, Yantai, China
| | - Min Chen
- College of Life Sciences, Yantai University, Yantai, China
| | - Lili Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- *Correspondence: Lili Li,
| |
Collapse
|
19
|
Zanetta P, Ormelli M, Amoruso A, Pane M, Azzimonti B, Squarzanti DF. Probiotics as Potential Biological Immunomodulators in the Management of Oral Lichen Planus: What's New? Int J Mol Sci 2022; 23:ijms23073489. [PMID: 35408849 PMCID: PMC8998608 DOI: 10.3390/ijms23073489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Oral lichen planus (OLP) is a T cell-mediated chronic inflammatory disorder with multifactorial aetiology and malignant transformation potential. Despite the treatments so far identified, new tailored and safe specific measures are needed. Recently, human microbiota imbalance has been linked to several immune-mediated diseases, opening new therapeutic perspectives for probiotics; besides their ability to directly interact with the host microbiota, they also display a strain-specific immune-modulatory effect. Thus, this non-systematic review aims to elucidate the molecular pathways underlying probiotic activity, mainly those of Lactobacilli and Bifidobacteria and their metabolites in OLP pathogenesis and malignant transformation, focusing on the most recent in vitro and in vivo research evidence. Findings related to their activity in other immune-mediated diseases are here included, suggesting a probiotic translational use in OLP. Probiotics show immune-modulatory and microbiota-balancing activities; they protect the host from pathogens, hamper an excessive effector T cell response, reduce nuclear factor-kappa B (NF-kB) signalling and basal keratinocytes abnormal apoptosis, shifting the mucosal response towards the production of anti-inflammatory cytokines, thus preventing uncontrolled damage. Therefore, probiotics could be a highly encouraging prevention and immunotherapeutic approach for a safer and more sustainable OLP management.
Collapse
Affiliation(s)
- Paola Zanetta
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
| | - Margherita Ormelli
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
| | - Angela Amoruso
- Probiotical Research Srl, Via Mattei 3, 28100 Novara, Italy; (A.A.); (M.P.)
| | - Marco Pane
- Probiotical Research Srl, Via Mattei 3, 28100 Novara, Italy; (A.A.); (M.P.)
| | - Barbara Azzimonti
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
- Correspondence: (B.A.); (D.F.S.); Tel.: +39-0321-660-870 (B.A.)
| | - Diletta Francesca Squarzanti
- Laboratory of Applied Microbiology, Department of Health Sciences (DiSS), Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), School of Medicine, Università del Piemonte Orientale (UPO), Corso Trieste 15/A, 28100 Novara, Italy; (P.Z.); (M.O.)
- Correspondence: (B.A.); (D.F.S.); Tel.: +39-0321-660-870 (B.A.)
| |
Collapse
|