1
|
Mosiej W, Długosz E, Kruk M, Zielińska D. Immunomodulatory Properties of Live and Thermally-Inactivated Food-Origin Lactic Acid Bacteria-In Vitro Studies. Mol Nutr Food Res 2025:e70047. [PMID: 40166824 DOI: 10.1002/mnfr.70047] [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: 10/21/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
The study investigates the strain-specific immunomodulatory properties of live and thermally-inactivated (TI) lactic acid bacteria (LAB) derived from traditional Polish fermented foods, focusing on their potential as probiotics and postbiotics. LAB strains, known for their role in food fermentation, were assessed for their ability to influence cytokine production in THP-1 macrophages, maintain intestinal epithelial barrier integrity in Caco-2 monolayers, exhibit antioxidant activity, and produce specific organic acids and sugars. The research demonstrated that live LAB strains significantly upregulated the anti-inflammatory cytokine IL-10, particularly under inflammatory conditions, while TI strains exhibited notable antioxidant and anti-inflammatory properties. TI strains showed a greater ability to protect epithelial barrier function and reduce pro-inflammatory cytokine secretion than live strains, suggesting a promising role for postbiotics. The findings underscore the potential of LAB from fermented foods, demonstrating that postbiotic derivatives can differently influence inflammation compared to live bacteria, highlighting their potential as immune-enhancing agents, capable of modulating immune responses and offering therapeutic benefits against inflammation-related disorders. However, the limitations of in vitro models highlight the need for further in vivo and clinical studies to validate these effects and fully uncover the health benefits of these LAB strains for humans.
Collapse
Affiliation(s)
- Wioletta Mosiej
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Ewa Długosz
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Marcin Kruk
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Dorota Zielińska
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| |
Collapse
|
2
|
Kim MJ, Lee YJ, Hussain Z, Park H. Effect of Probiotics on Improving Intestinal Mucosal Permeability and Inflammation after Surgery. Gut Liver 2025; 19:207-218. [PMID: 39327843 PMCID: PMC11907258 DOI: 10.5009/gnl240170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 09/28/2024] Open
Abstract
Background/Aims We explored the mechanisms underlying the improvement of postoperative ileus (POI) following probiotic pretreatment. We assessed intestinal permeability, inflammation, tight junction (TJ) protein expression in the gut epithelium, and plasma interleukin (IL)-17 levels in a guinea pig model of POI. Methods Guinea pigs were divided into control, POI, and probiotic groups. The POI and probiotic groups underwent surgery, but the probiotic group received probiotics before the procedure. The ileum and proximal colon were harvested. Intestinal permeability was measured via horseradish peroxidase permeability. Inflammation was evaluated via leukocyte count in the intestinal wall muscle layer, and calprotectin expression in each intestinal wall layer was analyzed immunohistochemically. TJ proteins were analyzed using immunohistochemical staining, and plasma IL-17 levels were measured using an enzyme-linked immunosorbent assay. Results The POI group exhibited increased intestinal permeability and inflammation, whereas probiotic pretreatment reduced the extent of these POI-induced changes. Probiotics restored the expression of TJ proteins occludin and zonula occludens-1 in the proximal colon, which were increased in the POI group. Calprotectin expression significantly increased in the muscle layer of the POI group and was downregulated in the probiotic group; however, no distinct differences were observed between the mucosal and submucosal layers. Plasma IL-17 levels did not significantly differ among the groups. Conclusions Probiotic pretreatment may relieve POI by reducing intestinal permeability and inflammation and TJ protein expression in the gut epithelium. These findings suggest a potential therapeutic approach for POI management.
Collapse
Affiliation(s)
- Min-Jae Kim
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Lee
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Zahid Hussain
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
O'Grady K, Grabrucker AM. Metal Dyshomeostasis as a Driver of Gut Pathology in Autism Spectrum Disorders. J Neurochem 2025; 169:e70041. [PMID: 40108935 PMCID: PMC11923526 DOI: 10.1111/jnc.70041] [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: 10/01/2024] [Revised: 01/31/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Despite being classified as neurodevelopmental disorders, in recent years, there has been a growing interest in the association between autism spectrum disorders (ASDs) and gut pathology. This comprehensive and systematic review explores a potential mechanism underlying gut pathology in ASDs, including alterations in gut microbiota, intestinal permeability, immune dysregulation, and gastrointestinal (GI) symptoms. Specifically, it delves into the role of toxic and essential metals and their interplay, affecting the development and function of the GI tract. The review also discusses the potential implications of this gut pathology in the development and management of ASDs. Studies have shown that heavy metal exposure, whether through environmental sources or dietary intake, can disrupt the delicate balance of trace elements in the gut. This disruption can adversely affect zinc homeostasis, potentially exacerbating gut pathology in individuals with ASDs. The impaired zinc absorption resulting from heavy metal exposure may contribute to the immune dysregulation, oxidative stress, and inflammation observed in the gut of individuals with ASDs. By shedding light on the multifaceted nature of gut pathology, including the impact of metal dyshomeostasis as a non-genetic factor in ASD, this review underscores the significance of the gut-brain axis in the etiology and management of ASDs.
Collapse
Affiliation(s)
- Katelyn O'Grady
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
4
|
Ekwudo MN, Gubert C, Hannan AJ. The microbiota-gut-brain axis in Huntington's disease: pathogenic mechanisms and therapeutic targets. FEBS J 2025; 292:1282-1315. [PMID: 38426291 PMCID: PMC11927060 DOI: 10.1111/febs.17102] [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: 10/13/2023] [Revised: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Huntington's disease (HD) is a currently incurable neurogenerative disorder and is typically characterized by progressive movement disorder (including chorea), cognitive deficits (culminating in dementia), psychiatric abnormalities (the most common of which is depression), and peripheral symptoms (including gastrointestinal dysfunction). There are currently no approved disease-modifying therapies available for HD, with death usually occurring approximately 10-25 years after onset, but some therapies hold promising potential. HD subjects are often burdened by chronic diarrhea, constipation, esophageal and gastric inflammation, and a susceptibility to diabetes. Our understanding of the microbiota-gut-brain axis in HD is in its infancy and growing evidence from preclinical and clinical studies suggests a role of gut microbial population imbalance (gut dysbiosis) in HD pathophysiology. The gut and the brain can communicate through the enteric nervous system, immune system, vagus nerve, and microbiota-derived-metabolites including short-chain fatty acids, bile acids, and branched-chain amino acids. This review summarizes supporting evidence demonstrating the alterations in bacterial and fungal composition that may be associated with HD. We focus on mechanisms through which gut dysbiosis may compromise brain and gut health, thus triggering neuroinflammatory responses, and further highlight outcomes of attempts to modulate the gut microbiota as promising therapeutic strategies for HD. Ultimately, we discuss the dearth of data and the need for more longitudinal and translational studies in this nascent field. We suggest future directions to improve our understanding of the association between gut microbes and the pathogenesis of HD, and other 'brain and body disorders'.
Collapse
Affiliation(s)
- Millicent N Ekwudo
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Australia
| |
Collapse
|
5
|
Wu Y, Zhang X, Wang GQ, Jiao Y. Clinical significance of perioperative probiotic intervention on recovery following intestinal surgery. World J Gastrointest Surg 2025; 17:97503. [DOI: 10.4240/wjgs.v17.i2.97503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/03/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Restoring the balance of gut microbiota has emerged as a critical strategy in treating intestinal disorders, with probiotics playing a pivotal role in maintaining bacterial equilibrium. Surgical preparations, trauma, and digestive tract reconstruction associated with intestinal surgeries often disrupt the intestinal flora, prompting interest in the potential role of probiotics in postoperative recovery. Lan et al conducted a prospective randomized study on 60 patients with acute appendicitis, revealing that postoperative administration of Bacillus licheniformis capsules facilitated early resolution of inflammation and restoration of gastrointestinal motility, offering a novel therapeutic avenue for accelerated postoperative recovery. This editorial delves into the effects of perioperative probiotic supplementation on physical and intestinal recovery following surgery. Within the framework of enhanced recovery after surgery, the exploration of new probiotic supplementation strategies to mitigate surgical complications and reshape gut microbiota is particularly intriguing.
Collapse
Affiliation(s)
- Yang Wu
- Department of Nephrology, Jilin People’s Hospital, Jilin 132000, Jilin Province, China
| | - Xin Zhang
- Department of Nephrology, Jilin People’s Hospital, Jilin 132000, Jilin Province, China
| | - Guan-Qiao Wang
- Department of Abdominal Tumor Surgery, Jilin Cancer Hospital, Changchun 130000, Jilin Province, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
6
|
Dong Y, Zheng Y, Liu H, Wang Y, Cui J, Wu Y, Yan L, Miao Z, Han M, Huang C, Li P, Su Y, Shen Y, Zhang J, Yuan J, Zhang B, Li J. Effects of high stocking density on the growth performance, intestinal health and bile salts composition of broiler chickens. Front Microbiol 2025; 16:1542059. [PMID: 40071203 PMCID: PMC11895766 DOI: 10.3389/fmicb.2025.1542059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction The intestinal dysfunction plays an important role in the decreased growth performance of broiler chickens under high stocking density. Gut microbiota plays an important role in maintaining intestinal health. However, the modulation pathway of gut microbiota by regulating the intestinal barrier and histomorphology remains unknown. Methods One hundred and forty-four male Arbor Acres broilers (22-d-old) with similar weight were randomly assigned to two treatments: a high (HSD, 20 broilers/m2) or low stocking density treatment (LSD, 14 broilers/m2), with six replicates per treatment. The experimental period was 20 days, from 22 to 42 days of age. Results The final body weight at 42 days of age was lower in the HSD group (P = 0.0013) and average daily feed intake (P = 0.016) and weight gain of broilers from 22 to 42 days decreased (P = 0.012). In the HSD group on day 42, villus height and the ratio of villus height to the crypt depth in the ileum decreased (P < 0.05); mRNA expression of tight junction proteins, occludin (P < 0.01) and ZO1 (P < 0.05), were downregulated; whereas IL-6, TNFα, and NFκB p65 (P < 0.05) and IL-1β (P < 0.01) were upregulated. The HSD treatment increased the relative abundance of Lactobacillus (P = 0.045) and decreased that of Alistipes (P = 0.031). Cecal concentrations of acetic (P < 0.05) and butyric acids (P < 0.05) decreased. Gut metabolites co-metabolized by the host and gut microbiota were altered in the HSD group, with decreases in glycerophospholipid and tryptophan metabolites negatively correlated with Lactobacillus (P < 0.05). The metabolite content of conjugated bile acids decreased and free bile acids increased (P < 0.05) with HSD. Bile salt hydrolase (BSH) was increased in the intestine of HSD-treated broilers (P < 0.01). The total cholic acid content of the HSD group was lower in the jejunum and ileum (P < 0.05) but higher in the cecum than in the LSD group (P < 0.01). Conclusion HSD caused dysbiosis of the intestinal microbiota such as increased Lactobacillus, along with enhanced BSH activity and excessive unabsorbed free bile acids. This resulted in ileal epithelial cells damage, inflammation, decreased growth performance of broilers under high stocking densities.
Collapse
Affiliation(s)
- Yuanyang Dong
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Yuqi Zheng
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Haoyu Liu
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Yaru Wang
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Jiaqing Cui
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Yuan Wu
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Lei Yan
- Shandong New Hope Liuhe Group Co., Ltd., Qingdao, China
| | - Zhiqiang Miao
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Miaomiao Han
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Chenxuan Huang
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Peifeng Li
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Yuan Su
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Yiru Shen
- Chinese Academy of Agricultural Sciences Poultry Institute, Yangzhou, Jiangsu, China
| | - Junzhen Zhang
- Shanxi Agricultural University College of Animal Science, Taigu, China
| | - Jianmin Yuan
- China Agricultural University College of Animal Science and Technology, Beijing, China
| | - Bingkun Zhang
- China Agricultural University College of Animal Science and Technology, Beijing, China
| | - Jianhui Li
- Shanxi Agricultural University College of Animal Science, Taigu, China
| |
Collapse
|
7
|
Kacena C. Effects of the Curcuminoid and Non-Curcuminoid Compounds of Turmeric on the Gut Microbiome and Inflammation: Potential Use in the Treatment and Prevention of Disease. Nutr Rev 2025:nuae221. [PMID: 39873671 DOI: 10.1093/nutrit/nuae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
The gut microbiome is a complex system that directly interacts with and influences many systems in the body. This delicate balance of microbiota plays an important role in health and disease and is highly influenced by lifestyle factors and the surrounding environment. As further research emerges, understanding the full potential of the gut microbiome and the impact of using nutraceuticals to positively influence its function may open the door to greater therapeutic outcomes in the treatment and prevention of disease. Curcumin, a bioactive compound derived from the turmeric rhizome, has been studied in depth for its influence on human health as a potent anti-inflammatory and antioxidant properties. However, the therapeutic activity of curcumin is limited by its low oral bioavailability. While most available research has primarily focused on the curcuminoid compounds of turmeric, the non-curcuminoid compounds hold promise to offer therapeutic benefits while synergistically enhancing the bioavailability of curcumin and supporting the gut microbiome. This review summarizes current knowledge of the relationship between the gut and the various systems within the body, and how dysbiosis, or disruption in the gut microbial balance, leads to inflammation and increased risk of chronic disease. The review also summarizes recent research that focuses on the bioactivity of both the curcuminoid and non-curcuminoid compounds that comprise the whole turmeric root and their synergistic role in enhancing bioavailability to support a healthy gut microbiome and promising use in the treatment and prevention of disease.
Collapse
|
8
|
Wankhede NL, Kale MB, Kyada A, M RM, Chaudhary K, Naidu KS, Rahangdale S, Shende PV, Taksande BG, Khalid M, Gulati M, Umekar MJ, Fareed M, Kopalli SR, Koppula S. Sleep deprivation-induced shifts in gut microbiota: Implications for neurological disorders. Neuroscience 2025; 565:99-116. [PMID: 39622383 DOI: 10.1016/j.neuroscience.2024.11.070] [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: 07/31/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sleep deprivation is a prevalent issue in contemporary society, with significant ramifications for both physical and mental well-being. Emerging scientific evidence illuminates its intricate interplay with the gut-brain axis, a vital determinant of neurological function. Disruptions in sleep patterns disturb the delicate equilibrium of the gut microbiota, resulting in dysbiosis characterized by alterations in microbial composition and function. This dysbiosis contributes to the exacerbation of neurological disorders such as depression, anxiety, and cognitive decline through multifaceted mechanisms, including heightened neuroinflammation, disturbances in neurotransmitter signalling, and compromised integrity of the gut barrier. In response to these challenges, there is a burgeoning interest in therapeutic interventions aimed at restoring gut microbial balance and alleviating neurological symptoms precipitated by sleep deprivation. Probiotics, dietary modifications, and behavioural strategies represent promising avenues for modulating the gut microbiota and mitigating the adverse effects of sleep disturbances on neurological health. Moreover, the advent of personalized interventions guided by advanced omics technologies holds considerable potential for tailoring treatments to individualized needs and optimizing therapeutic outcomes. Interdisciplinary collaboration and concerted research efforts are imperative for elucidating the underlying mechanisms linking sleep, gut microbiota, and neurological function. Longitudinal studies, translational research endeavours, and advancements in technology are pivotal for unravelling the complex interplay between these intricate systems.
Collapse
Affiliation(s)
- Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences Marwadi University, Rajkot 360003, Gujarat, India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sandip Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
9
|
Indrio F, Salatto A. Gut Microbiota-Bone Axis. ANNALS OF NUTRITION & METABOLISM 2025:1-10. [PMID: 39848230 DOI: 10.1159/000541999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/11/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Knowledge of the complex interplay between gut microbiota and human health is gradually increasing as it has just recently been a field of such great interest. SUMMARY Recent studies have reported that communities of microorganisms inhabiting the gut influence the immune system through cellular responses and shape many physiological and pathophysiological aspects of the body, including muscle and bone metabolism (formation and resorption). Specifically, the gut microbiota affects skeletal homeostasis through changes in host metabolism, the immune system, hormone secretion, and the gut-brain axis. The major role on gut-bone axis is due to short-chain fatty acids (SCFAs). They have the ability to influence regulatory T-cell (Tregs) development and activate bone metabolism through the action of Wnt10. SCFA production may be a mechanism by which the microbial community, by increasing the serum level of insulin-like growth factor 1 (IGF-1), leads to the growth and regulation of bone homeostasis. A specific SCFA, butyrate, diffuses into the bone marrow where it expands Tregs. The Tregs induce production of the Wnt ligand Wnt10b by CD8+ T cells, leading to activation of Wnt signaling and stimulation of bone formation. At the hormonal level, the effect of the gut microbiota on bone homeostasis is expressed through the biphasic action of serotonin. Some microbiota, such as spore-forming microbes, regulate the level of serotonin in the gut, serum, and feces. Another group of bacterial species (Lactococcus, Mucispirillum, Lactobacillus, and Bifidobacterium) can increase the level of peripheral/vascular leptin, which in turn manages bone homeostasis through the action of brain serotonin.
Collapse
Affiliation(s)
- Flavia Indrio
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Alessia Salatto
- Department of Translational Medical Science, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
10
|
Barata P, Oliveira A, Soares R, Fernandes A. Gut Microbiota Is Not Significantly Altered by Radioiodine Therapy. Nutrients 2025; 17:395. [PMID: 39940254 PMCID: PMC11819986 DOI: 10.3390/nu17030395] [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: 12/14/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/14/2025] Open
Abstract
Purpose: Radiotherapy treatments are known to alter the gut microbiota. However, little is known regarding the effect of nuclear medicine treatments on gut microbiota, and it is established that nuclear medicine is inherently different from radiotherapy. To address this knowledge gap, we conducted a prospective study to identify changes in the gut microbiota of patients treated with [131I]NaI by comparing fecal samples before and after RAIT. Methods: Fecal samples of 64 patients (37 with thyroid cancer and 27 with hyperthyroidism) with indication for RAIT were collected 2 to 3 days before treatment and 8 to 10 days post-treatment. After DNA extraction, the gut microbiota's richness, diversity, and composition were analyzed by shotgun metagenomics. In addition, LEfSe was performed to compare compositional changes in specific bacteria. Results: Gut microbiome richness and diversity remained unchanged after RAIT, with few changes in its composition identified, especially in patients with hyperthyroidism. Conclusions: This study provides a conceptual and analytical basis for increasing our understanding of the effects of radiopharmaceuticals on gut microbiota. Our preliminary results indicate that RAIT, contrary to radiotherapy, does not cause major disruptions to the human gut microbiota.
Collapse
Affiliation(s)
- Pedro Barata
- RISE-Health, Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Fundação Ensino e Cultura Fernando Pessoa, Rua Carlos da Maia 296, 4200-150 Porto, Portugal
- Centro Hospitalar de Santo António, Unidade Local de Saúde de Santo António, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal
| | - Ana Oliveira
- Department of Nuclear Medicine, Centro Hospitalar e Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| | - Raquel Soares
- Department of Biomedicine, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Ana Fernandes
- RISE-Health, Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Fundação Ensino e Cultura Fernando Pessoa, Rua Carlos da Maia 296, 4200-150 Porto, Portugal
- Department of Nuclear Medicine, Centro Hospitalar e Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| |
Collapse
|
11
|
Goel A, Ncho CM, Jeong CM, Gupta V, Jung JY, Ha SY, Yang JK, Choi YH. Alleviation of Acute Heat Stress in Broiler Chickens by Dietary Supplementation of Polyphenols from Shredded, Steam-Exploded Pine Particles. Microorganisms 2025; 13:235. [PMID: 40005601 PMCID: PMC11858311 DOI: 10.3390/microorganisms13020235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Reducing the detrimental effects of heat stress (HS) in poultry is essential to minimize production losses. The present study evaluates the effects of dietary polyphenols prepared from underutilized wood byproducts on the growth, gut health, and cecal microbiota in broilers subjected to acute heat stress (AHS). One hundred eight one-day-old Indian River broilers were fed with 0%, 0.5%, or 1% polyphenols from shredded, steam-exploded pine particles (PSPP) in their diet. On the 37th day, forty birds were equally distributed among four groups containing either a control diet at thermoneutral temperatures (NT0%) or AHS temperatures with 0% (AHS0%), 0.5% (AHS0.5%), and 1% (AHS1%) PSPP-supplemented diets. The temperature in the NT room was maintained at 21.0 °C, while, in the AHS room, it was increased to 31 °C. AHS negatively influenced performance parameters and increased rectal temperature (RT) in broilers. The AHS0% group showed a higher expression of NOX4, HSP-70, and HSP-90 genes, while the expression was lower in PSPP-supplemented birds. In the jejunum, mRNA expression of SOD was increased in all the birds under AHS compared to NT. The expression of the CLDN1 and ZO2 genes was increased in AHS0%, while that of the ZO1 and MUC2 genes was increased in PSPP-supplemented birds. HS tends to increase TLR2 and TLR4 gene expression in chickens. The significantly modified genera were Bariatricus, Sporobacter, Sporanaerobacter, and Natranaerovirga. Concludingly, AHS negatively influences the performance parameters, RT, stress, gut-health-related genes, and pathogenic penetration, but PSPP supplementation reduces its bad impact by overcoming the stress and gut-health-related genes, increasing favorable bacterial abundance and reducing pathogenic penetration in chickens.
Collapse
Affiliation(s)
- Akshat Goel
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (A.G.); (C.-M.N.); (C.-M.J.); (V.G.)
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.-Y.J.); (J.-K.Y.)
| | - Chris-Major Ncho
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (A.G.); (C.-M.N.); (C.-M.J.); (V.G.)
| | - Chae-Mi Jeong
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (A.G.); (C.-M.N.); (C.-M.J.); (V.G.)
- Division of Applied Life Sciences (BK21 Four Program), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Vaishali Gupta
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (A.G.); (C.-M.N.); (C.-M.J.); (V.G.)
- Division of Applied Life Sciences (BK21 Four Program), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ji-Young Jung
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.-Y.J.); (J.-K.Y.)
- Department of Environmental Materials Science, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Si-Young Ha
- Department of Environmental Materials Science, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Jae-Kyung Yang
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.-Y.J.); (J.-K.Y.)
- Department of Environmental Materials Science, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Yang-Ho Choi
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Republic of Korea; (A.G.); (C.-M.N.); (C.-M.J.); (V.G.)
- Institute of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.-Y.J.); (J.-K.Y.)
- Division of Applied Life Sciences (BK21 Four Program), Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
12
|
Tu M, Cai G, Ma L, Yan L, Wang T, Shi Z, Wang C, Chen Z. Effects of Different Levels of Lycium barbarum Flavonoids on Growth Performance, Immunity, Intestinal Barrier and Antioxidant Capacity of Meat Ducks. Antioxidants (Basel) 2025; 14:67. [PMID: 39857401 PMCID: PMC11761579 DOI: 10.3390/antiox14010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Background: In vitro findings on the biological functions of Lycium barbarum flavonoids (LBFs) as feed additives are limited. This study aimed to explore the effects of different concentrations of LBFs on the growth performance, immune function, intestinal barrier, and antioxidant capacity of meat ducks. A total of 240 one-day-old male meat ducks were randomly allocated to four groups, each receiving a basal diet supplemented with 0 (control), 250, 500, or 1000 mg/kg of LBFs for 42 d. Results: The results showed that dietary supplementation with 500 mg/kg of LBFs resulted in a significant increase in average daily feed intake, body weight, average daily gain, and feed conversion ratio. Dietary supplementation with 500 or 1000 mg/kg of LBFs resulted in significant decreases in serum levels of D-lactic acid and lipopolysaccharide. Dietary supplementation with 500 mg/kg LBFs significantly decreased diamine oxidase activity and enhanced the activities of catalase, total antioxidant capacity, and glutathione peroxidase in the jejunal mucosa, as well as the activity of total superoxide dismutase and the content of glutathione in the ileal mucosa, while significantly lowering the content of malondialdehyde in the ileal mucosa. Dietary supplementation with 500 mg/kg LBFs significantly up-regulated the mRNA expression of genes associated with intestinal barrier function and antioxidant capacity in the jejunal and ileal mucosa, as well as the protein expression of these antioxidant genes, and led to a significant reduction in the mRNA expression of pro-apoptotic and inflammatory-related genes. Conclusions: The addition of LBFs to the diet improved the growth performance, intestinal barrier function, immune response, and antioxidant capacity of the ducks, which may be closely associated with the activation of the Nrf2 signaling pathway and the inhibition of the NF-κB signaling pathway. The optimal dietary inclusion level of LBFs in ducks was 500 mg/kg.
Collapse
Affiliation(s)
- Minhang Tu
- College of Animal Sciences and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.T.); (G.C.); (L.M.); (T.W.)
| | - Gentan Cai
- College of Animal Sciences and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.T.); (G.C.); (L.M.); (T.W.)
| | - Longfei Ma
- College of Animal Sciences and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.T.); (G.C.); (L.M.); (T.W.)
| | - Leyan Yan
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (L.Y.); (Z.S.)
- Integrated Crop-Livestock Systems Key Laboratory, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| | - Tian Wang
- College of Animal Sciences and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.T.); (G.C.); (L.M.); (T.W.)
| | - Zhendan Shi
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (L.Y.); (Z.S.)
- Integrated Crop-Livestock Systems Key Laboratory, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| | - Chao Wang
- College of Animal Sciences and Technology, Nanjing Agricultural University, Nanjing 210095, China; (M.T.); (G.C.); (L.M.); (T.W.)
| | - Zhe Chen
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (L.Y.); (Z.S.)
- Integrated Crop-Livestock Systems Key Laboratory, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| |
Collapse
|
13
|
Ahrend H, Buchholtz A, Stope MB. Microbiome and Mucosal Immunity in the Intestinal Tract. In Vivo 2025; 39:17-24. [PMID: 39740876 PMCID: PMC11705094 DOI: 10.21873/invivo.13801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025]
Abstract
The human bowel is exposed to numerous biotic and abiotic external noxious agents. Accordingly, the digestive tract is frequently involved in malfunctions within the organism. Together with the commensal intestinal flora, it regulates the immunological balance between inflammatory defense processes and immune tolerance. Pathological changes in this system often cause chronic inflammatory bowel diseases including Crohn's disease and ulcerative colitis. This review article highlights the complex interaction between commensal microorganisms, the intestinal microbiome, and the intestinal epithelium-localized local immune system. The main functions of the human intestinal microbiome include (i) protection against pathogenic microbial colonization, (ii) maintenance of the barrier function of the intestinal epithelium, (iii) degradation and absorption of nutrients and (iv) active regulation of the intestinal immunity. The local intestinal immune system consists primarily of macrophages, antigen-presenting cells, and natural killer cells. These cells regulate the commensal intestinal microbiome and are in turn regulated by signaling factors of the epithelial cells and the microbiome. Deregulated immune responses play an important role and can lead to both reduced activity of the commensal microbiome and pathologically increased activity of harmful microorganisms. These aspects of chronic inflammatory bowel disease have become the focus of attention in recent years. It is therefore important to consider the immunological-microbial context in both the diagnosis and treatment of inflammatory bowel diseases. A promising holistic approach would include the most comprehensive possible diagnosis of the immune and microbiome status of the patient, both at the time of diagnostics and during therapy.
Collapse
Affiliation(s)
- Hannes Ahrend
- Department of Medicine, Israelite Hospital Hamburg, Hamburg, Germany
| | - Anja Buchholtz
- Department of Medicine, Israelite Hospital Hamburg, Hamburg, Germany
| | - Matthias B Stope
- Department of Gynecology and Gynecological Oncology, Research Laboratories, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
14
|
Rafieian-Naeini HR, Ko H, Goo D, Choppa VSR, Gudidoddi SR, Katha HR, Kim WK. Synergistic impact of Salmonella typhimurium and Eimeria spp. coinfection on turkey poults: Growth performance, salmonella colonization, and ceca microbiota insights. Poult Sci 2025; 104:104568. [PMID: 39647353 PMCID: PMC11667029 DOI: 10.1016/j.psj.2024.104568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024] Open
Abstract
Salmonella contamination in poultry products is a significant concern due to its potential to cause severe economic losses and public health problems. On the other side, coccidiosis is induced by Eimeria (E.) species. involves the destruction of host intestinal epithelial cells and subsequent invasion of pathogens, resulting in performance reduction and enhanced pathogen infection in poultry and economic losses for the poultry industry. A study was conducted to evaluate the impact of Eimeria infection and Salmonella typhimurium (ST) on growth performance, Salmonella colonization, and ceca microbiota in turkey poult. A total of 420 one-day-old male turkey poults were randomly allocated into six treatments, with five replicated cages for each treatment, over a 21-day experimental period. The study followed a 2 × 3 factorial design. Treatments consisted of NC, negative control without any challenge; T1, challenged with 8000 oocysts of E. meleagrimitis and E. adenoeides at d 8; T2, challenged with 16,000 oocysts of E. meleagrimitis and E. adenoeides at d 8; T3, challenged with nalidixic acid-resistant Salmonella typhimurium (ST) at d 0; T4, challenged with ST at d 0 and 8000 oocysts of E. meleagrimitis and E. adenoeides at d 8; T5, challenged with ST at d 0 and 16,000 oocysts of E. meleagrimitis and E. adenoeides at d 8. The Eimeria challenge groups significantly reduced the BW compared to the non-challenge group (P < 0.001). The challenged groups decreased FI during 9-14 days of age (P < 0.01). Salmonella typhimurium did not affect BW entire trial period (P > 0.05). Gut permeability (GP) increased in the challenge groups compared to the NC group (P < 0.001). Both ST and Eimeria significantly decreased superoxide dismutase (SOD) in the liver (P < 0.01). The challenge groups had lower villus height (VH) and higher crypt depth (CD) compared to the NC group, resulting in decreased VH:CD ratio in the duodenum and jejunum (P < 0.01). The groups T1, T2, and T4 had significantly higher fat deposition than the NC group (P < 0.05). The coinfected groups (T4 and T5) had higher salmonella colonization in the spleen compared to the ST-infected group (T3, P < 0.05). The ST challenge significantly decreased alpha diversity, including pielou evenness and Shannon entropy (P < 0.05). The Proteobacteria phylum and Enterobacteriaceae family significantly increased in T5 compared to the NC, T1, T2, and T3 (P < 0.05). In conclusion, Eimeria infection negatively impacted growth, gut health, intestine barrier integrity, and histology, while Salmonella had a milder effect on performance. Coinfection with Salmonella and Eimeria spp. led to changes in gut microbiota and increased liver Salmonella colonization and fat deposition in turkey poults.
Collapse
Affiliation(s)
| | - Hanseo Ko
- Department of Poultry Science, University of Georgia, Athens, GA 30602-2772, USA
| | - Doyun Goo
- Department of Poultry Science, University of Georgia, Athens, GA 30602-2772, USA
| | | | | | - Hemanth Reddy Katha
- Department of Poultry Science, University of Georgia, Athens, GA 30602-2772, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602-2772, USA.
| |
Collapse
|
15
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
16
|
Chen N, Yao P, Farid MS, Zhang T, Luo Y, Zhao C. Effect of bioactive compounds in processed Camellia sinensis tea on the intestinal barrier. Food Res Int 2025; 199:115383. [PMID: 39658174 DOI: 10.1016/j.foodres.2024.115383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
The human intestinal tract plays a pivotal role in safeguarding the body against noxious substances and microbial pathogens by functioning as a barrier. This barrier function is achieved through the combined action of physical, chemical, microbial, and immune components. Tea (Camellia sinensis) is the most widely consumed beverage in the world, and it is consumed and appreciated in a multitude of regions across the globe. Tea can be classified into various categories, including green, white, yellow, oolong, black, and dark teas, based on the specific processing methods employed. In recent times, there has been a notable surge in scientific investigation into the various types of tea. The recent surge in research on tea can be attributed to the plethora of bioactive compounds it contains, including polyphenols, polysaccharides, pigments, and theanine. The processing of different teas affects the active ingredients to varying degrees, resulting in a range of chemical reactions and the formation of different types and quantities of ingredients. The bioactive compounds present in tea are of great importance for the maintenance of the integrity of the intestinal barrier, operating through a variety of mechanisms. This literature review synthesizes scientific studies on the impact of the primary bioactive compounds and different processing methods of tea on the intestinal barrier function. This review places particular emphasis on the exploration of the barrier repair and regulatory effects of these compounds, including the mitigation of damage to different barriers following intestinal diseases. Specifically, the active ingredients in tea can alleviate damage to physical barriers and chemical barriers by regulating barrier protein expression. At the same time, they can also maintain the stability of immune and biological barriers by regulating the expression of inflammatory factors and the metabolism of intestinal flora. This investigation can establish a strong theoretical foundation for the future development of innovative tea products.
Collapse
Affiliation(s)
- Nan Chen
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Peng Yao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | | | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Yangchao Luo
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, United States.
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
17
|
Li X, Li C, Wu P, Zhang L, Zhou P, Ma X. Recent status and trends of innate immunity and the gut-kidney aixs in IgAN: A systematic review and bibliometric analysis. Int Immunopharmacol 2024; 143:113335. [PMID: 39423662 DOI: 10.1016/j.intimp.2024.113335] [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: 05/26/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND There is a significant global demand for precise diagnosis and effective treatment of IgA nephropathy (IgAN), with innate immunity, particularly the complement system, exerting a profound influence on its pathogenesis. Additionally, the gut-kidney axis pathway is vital in the emergence and development of IgAN. METHODS We conducted a comprehensive search in the Web of Science database, spanning from January 1, 2000 to December 18, 2023. The gathered literature underwent a visual examination through CiteSpace, VOSviewer, and Scimago Graphica to delve into authors, nations, organizations, key terms, and other pertinent elements. RESULT Between 2000 and 2023, a total of 720 publications were identified, out of which 436 publications underwent screening for highly relevant literature analysis. The average annual number of articles focusing on IgAN, innate immunity, and the gut-kidney axis is approximately 31, with an upward trend observed. In terms of research impact encompassing publication count and authorship, the United States emerged as the leading contributor. Prominent keywords included "complement", "activation", "microbe", "gut-kidney axis", "C4d deposition", "alternative pathway" and "B cells" along with other prospective hot topics. CONCLUSION The correlation between IgAN and innate immunity is a focal point in current scientific research. Recent literature underscores the significance of the gut-kidney axis, where intestinal microorganisms and metabolites may influence IgAN. The complement system, a key component of innate immunity, also has a crucial function.Advancements in prevention, diagnosis, and treatment hinge on unraveling this intricate relationship.
Collapse
Affiliation(s)
- Xun Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Chengni Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Peiwen Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Lifang Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Ping Zhou
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| | - Xin Ma
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| |
Collapse
|
18
|
Zhong Y, Chang X, Zhao Z, Zheng L, Kuang G, Li P, Liu C, Fan Y, Liang Z, Zhuang K, Xie Q, Liu Y. Bacteroides fragilis capsular polysaccharide A ameliorates ulcerative colitis in rat by recovering intestinal barrier integrity and restoring gut microbiota. Front Pharmacol 2024; 15:1402465. [PMID: 39776580 PMCID: PMC11703662 DOI: 10.3389/fphar.2024.1402465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 10/02/2024] [Indexed: 01/11/2025] Open
Abstract
Bacteroides fragilis (B. fragilis) is a Gram-negative, obligate anaerobic, commensal bacterium residing in the human gut and holds therapeutic potential for ulcerative colitis (UC). Previous studies have indicated that capsular polysaccharide A (PSA) of B. fragilis is a crucial component for its effectiveness, possessing various biological activities such as anti-inflammatory, anti-tumor, and immune-modulating effects. We previously isolated and characterized the B. fragilis strain ZY-312 from the feces of a healthy breastfed infant, and extracted its PSA, named TP2. In this study, we explored the impact of TP2 on colonic inflammation and delved into its potential mechanisms. Initially, we used 2,4,6-trinitrobenzenesulfonic acid (TNBS) to induce colitis in rats and found that TP2 treatment significantly ameliorated TNBS-induced weight loss, increased clinical scores, extensive ulcers, and intestinal epithelial damage in UC rats. Further analysis revealed that TP2 effectively restored the intestinal barrier integrity in UC rats by regulating the expression of Muc-2, tight junction proteins (ZO-1, occludin, claudin-1, and claudin-2), as well as apoptosis-related proteins Bcl-2, BAX, and Cleaved-Caspase-3. Additionally, TP2 suppressed the expression of pro-inflammatory cytokines TNF-α, IL-1β, IL-6, and IL23, while promoting the secretion of anti-inflammatory cytokines IL-10 and IL-22, thereby inhibiting the occurrence of inflammation. TP2 also downregulated the phosphorylation levels of AKT and PI3K, effectively inhibiting the abnormal activation of the PI3K/AKT signaling pathway. More interestingly, 16S rRNA sequencing results showed that TP2 restored the ecological imbalance of the rat intestinal microbiota, with an increase in beneficial bacteria such as Lactobacillus and Limosilactobacillus observed in the treatment group. In conclusion, TP2 through the regulation of intestinal barrier-related cells and proteins, inhibition of apoptosis, modulation of inflammation-related cytokine levels, and control of abnormal activation of the PI3K/AKT signaling pathway, restores intestinal barrier integrity. Additionally, by reshaping the ecological imbalance of the gut microbiota, TP2 ultimately alleviates ulcerative colitis in rats.
Collapse
Affiliation(s)
- Yijia Zhong
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiujuan Chang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Zihan Zhao
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Lijun Zheng
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Gaobo Kuang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Ping Li
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | | | - Yuqin Fan
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhixuan Liang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ke Zhuang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qiuling Xie
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yangyang Liu
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| |
Collapse
|
19
|
Jiang J, Chen D, Yu B, He J, Yu J, Mao X, Huang Z, Luo Y, Luo J, Zheng P. Lactic Acid and Glutamine Have Positive Synergistic Effects on Growth Performance, Intestinal Function, and Microflora of Weaning Piglets. Animals (Basel) 2024; 14:3532. [PMID: 39682497 DOI: 10.3390/ani14233532] [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: 10/22/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
The objective of this study was to evaluate the effects of dietary addition of lactic acid and glutamine, and their interactions, on growth performance, nutrient digestibility, digestive enzyme activity, intestinal barrier functions, microflora, and expressions of intestinal development-related genes of weaning piglets. Ninety-six 24-day-old weaning piglets (Duroc × Landrace × Yorkshire, weaned at 21 ± 1 d and fed the basal diet for a 3 d adaptation period) with initial body weight of 7.24 ± 0.09 kg were randomly assigned to one of four dietary treatments with six replicates per treatment and four pigs per replicate in a 2 × 2 factorial treatment arrangements: (1) CON (a 2-period basal diet; control), (2) LS (supplemented with 2% lactic acid), (3) GS (supplemented with 1% glutamine), and (4) LGS (supplemented with 2% lactic acid and 1% glutamine). The study lasted for 28 d. On days 25-28, fresh fecal samples were collected to evaluate apparent total tract digestibility (ATTD) of nutrients. After 28 d, one weaning pig per pen was euthanized, and physiological samples obtained. Results showed that the supplementation of lactic acid improved the ADFI of the pigs (p < 0.05), while the pigs fed the glutamine diet had a greater ADFI and higher G/F (p < 0.05), and there were significant interactive effects between lactic acid and glutamine on the ADFI and G/F of the pigs (p < 0.05). The ATTD of CP and ash for pigs fed with lactic acid was significantly enhanced, and pigs fed the glutamine diet had greater ATTD of CP and ash (p < 0.05), while there were significant interactive effects between lactic acid and glutamine on the ATTD of CP and ash of the pigs (p < 0.05). Pigs fed with lactic acid exhibited greater activity of α-amylase and lipase (p < 0.05); moreover, the activity of lipase in the pigs showed a significant interactive effect between lactic acid and glutamine (p < 0.05). There was a greater villus height and villus height to crypt depth ratio in pigs fed with lactic acid (p < 0.05), and the villus height to crypt depth ratio of pigs fed with glutamine was greater (p < 0.05). There were greater GLUT2, IGF-1, TGF-β2, OCLN, and ZO-1 mRNA levels in pigs fed with lactic acid (p < 0.05), and the supplementation of glutamine increased SGLT1, GLUT2, PepT1, IGF-1, IGF-1R, TGFβ-2, GLP-2, and OCLN mRNA levels (p < 0.05), Additionally, expressions of SGLT1, GLUT2, PepT1, IGF-1, IGF-1R, TGFβ-2, GLP-2, CLDN-2, OCLN, and ZO-1 mRNA levels of pigs showed a positive interactive effect between lactic acid and glutamine (p < 0.05). Supplementation of lactic acid significantly increased the populations of Bifidobacterium in cecal digesta, Lactobacillus in colonic digesta, and the content of butyric acid in colonic digesta (p < 0.05). In addition, there were significant interactive effects between lactic acid and glutamine on populations of Bifidobacterium in cecal digesta, Lactobacillus in colonic digesta, and the content of acetic acid, butyric acid, and total VFAs in cecal digesta of the pigs (p < 0.05). Collectively, the current results indicate that dietary supplementation with lactic acid and glutamine had a positive synergistic effect on weaning pigs, which could improve growth performance through promoting the development of the small intestine, increasing digestive and barrier function, and regulating the balance of microflora in pigs, and which might be a potential feeding additive ensemble to enhance the health and growth of weaning piglets in the post-antibiotic era.
Collapse
Affiliation(s)
- Junjie Jiang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
20
|
Kishawy AT, Abd El-Wahab RA, Eldemery F, Abdel Rahman MMI, Altuwaijri S, Ezz-Eldin RM, Abd-Allah EM, Zayed S, Mulla ZS, El Sharkawy RB, Badr S, Youssef W, Ibrahim D. Insights of early feeding regime supplemented with glutamine and various levels of omega-3 in broiler chickens: growth performance, muscle building, antioxidant capacity, intestinal barriers health and defense against mixed Eimeria spp infection. Vet Q 2024; 44:1-20. [PMID: 38961536 PMCID: PMC11225632 DOI: 10.1080/01652176.2024.2373287] [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: 02/05/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024] Open
Abstract
Early nutritional management approach greatly impacts broilers' performance and resistance against coccidiosis. The current study explored the impact of post-hatch feeding with a combination of glutamine (Glut) and different levels of omega-3 on broiler chickens' growth performance, muscle building, intestinal barrier, antioxidant ability and protection against avian coccidiosis. A total of six hundred Cobb 500 was divided into six groups: first group (fed basal diet and unchallenged (control) and challenged (negative control, NC) groups were fed a basal diet without additives, and the other groups were infected with Eimeria spp and supplemented with 1.5% Glut alone or with three different levels of omega-3 (0.25, 0.5 and 1%) during the starter period. Notable improvement in body weight gain was observed in the group which fed basal diet supplemented with glut and 1% omega 3 even after coccidia infection (increased by 25% compared challenged group) while feed conversion ratio was restored to control. Myogeneis was enhanced in the group supplemented with Glut and omega-3 (upregulation of myogenin, MyoD, mechanistic target of rapamycin kinase and insulin like growth factor-1 and downregulating of myostatin genes). Groups supplemented with Glut and higher levels of omega-3 highly expressed occluding, mucin-2, junctional Adhesion Molecule 2, b-defensin-1 and cathelicidins-2 genes. Group fed 1% Glut + omega-3 showed an increased total antioxidant capacity and glutathione peroxidase and super oxide dismutase enzymes activities with reduced levels of malondialdehyde, reactive oxygen species and H2O2. Post-infection, dietary Glut and 1% omega-3 increased intestinal interleukin-10 (IL) and secretory immunoglobulin-A and serum lysozyme, while decreased the elevated inflammatory mediators comprising interleukin IL-6, tumor necrosis factor-alpha, nitric oxide (NO) and inducible NO synthase. Fecal oocyst excretion and lesions score severity were lowered in the group fed 1% Glut and omega 3. Based on these findings, dietary Glut and omega-3 supplementation augmented restored overall broilers' performance after coccidial challenge.
Collapse
Affiliation(s)
- Asmaa T.Y Kishawy
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Reham A. Abd El-Wahab
- Biochemistry Department, Animal Health Research Institute (AHRI), Mansoura Branch, Agriculture Research Center (ARC), Giza, Egypt
| | - Fatma Eldemery
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | - Saleh Altuwaijri
- Department of Pathology and laboratory diagnosis, College of Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Rasha M.M. Ezz-Eldin
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ehab M. Abd-Allah
- Veterinary Educational Hospital, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Shimaa Zayed
- Biochemistry Department, Animal Health Research Institute (AHRI), Mansoura Branch, Agriculture Research Center (ARC), Giza, Egypt
| | - Zohair S. Mulla
- Department of Public Health, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudia Arabia
| | - Rasha B. El Sharkawy
- Department of Clinical Pathology, Zagazig Branch, Animal Health Research Institute (AHRI), Agriculture Research Center, Zagazig, Egypt
| | - Shereen Badr
- Department of Clinical Pathology, Animal Health Research Institute (AHRI), Mansoura Branch, Agricultural Research Center (ARC), Giza, Egypt
| | - Wessam Youssef
- Department of Biotechnology, Animal Health Research Institute (AHRI), Giza, Egypt
| | - Doaa Ibrahim
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
21
|
Goo D, Lee J, Paneru D, Sharma MK, Rafieian-Naeini HR, Mahdavi FS, Gyawali I, Gudidoddi SR, Han G, Kim WK. Effects of branched-chain amino acid imbalance and dietary valine and isoleucine supplementation in modified corn-soybean meal diets with corn distillers dried grains with solubles on growth performance, carcass quality, intestinal health, and cecal microbiome in Cobb 500. Poult Sci 2024; 103:104483. [PMID: 39510006 PMCID: PMC11577229 DOI: 10.1016/j.psj.2024.104483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
One important feature of corn distillers dried grains with solubles (DDGS) is its high leucine:lysine ratio, which can inhibit chicken growth by causing branched-chain amino acid (BCAA) antagonism. The current study was conducted to investigate the effects of BCAA imbalance of inclusion of DDGS and whether additional dietary valine and isoleucine could alleviate the negative effects in broilers. A total of 640 0-d-old male Cobb 500 broilers were allocated into 4 treatments with 8 replicates and reared until d 42. The four different dietary groups were as follows: 1) control (CON) group (corn-soybean meal-based diet); 2) 30% DDGS (30D) group (replacing soybean meal with 30% DDGS); 3) 30D + additional valine and isoleucine (30DB) group; and 4) the group of 30DB + additional valine and isoleucine to provide the same leucine:valine and leucine:isoleucine ratios as the CON group (30DBB). The analyzed leucine:lysine ratios of the CON group were 1.36/1.41/1.46 (starter/grower/finisher phase), whereas the average leucine:lysine ratios of the 30% DDGS groups were 1.61/1.70/1.78 (starter/grower/finisher phase). The 30% DDGS groups (30D, 30DB, and 30DBB) negatively affected body weight (BW) from d 7 to 42 and BW gain (BWG), feed intake, carcass weight, breast muscle weight, and jejunal and ileal villus height:crypt depth during the overall period (d 0 to 42) (P < 0.05). Furthermore, the 30% DDGS groups significantly altered expression levels of jejunal tight junction proteins, breast muscle mechanistic target of rapamycin (mTOR) pathway-related genes, BCAA catabolism genes, and AA transporters compared to the CON (P < 0.01). The 30% DDGS groups showed differences in beta-diversity indices compared to the CON group (P < 0.05). The 30DBB group showing the lowest d 21 and 42 BW and overall BWG had the largest differences compared to the CON group in most measurements. In conclusion, excessive replacement of soybean meal with DDGS can significantly increase leucine levels, which may negatively affect chicken growth. Additionally, inappropriate ratios of valine and isoleucine can further decrease growth performance.
Collapse
Affiliation(s)
- Doyun Goo
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Jihwan Lee
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Deependra Paneru
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Milan K Sharma
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | | | - Fatemeh S Mahdavi
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Ishwari Gyawali
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | | | - Gippeum Han
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA, United States.
| |
Collapse
|
22
|
Wang X, Yao DS, Xu L, Yan DM, Zhao Y, Peng JH, Fu QL, Hu YY, Feng Q. Jianpi Huoxue Decoction Ameliorates Alcohol-associated Liver Disease by Improving Intestinal Barrier Function in Rats. Curr Med Sci 2024; 44:1241-1248. [PMID: 39617866 DOI: 10.1007/s11596-024-2955-2] [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: 04/09/2024] [Accepted: 10/08/2024] [Indexed: 12/24/2024]
Abstract
OBJECTIVE Jianpi huoxue decoction (JHD), a Chinese herbal formula, is commonly used for treating alcohol-associated liver disease (ALD). This study aimed to investigate the mechanism by which JHD affects intestinal barrier function in ALD rats. METHODS The Sprague-Dawley rats were randomly divided into three groups: control group, model group and JHD group. They were pair-fed a modified Lieber-DeCarli liquid diet containing alcohol (model group, n=10; JHD group, n=10) or isocaloric maltose dextrin (control group, n=10) for 6 weeks. After 3 weeks of feeding, the mice in the JHD group were given JHD (10 mL/kg/day) by gavage for 3 weeks, and those in the control and model groups received equal amounts of double-distilled water for the same period of time. Afterwards, all the rats were given lipopolysaccharide (LPS) by gavage and sacrificed 3.5 h later. LPS levels were measured in the portal blood to evaluate gut leakage. Transmission electron microscopy (TEM) was used to observe ultrastructural changes in the intestinal tract. Adherens junction (AJ) and tight junction (TJ) proteins were detected by Western blotting, immunofluorescence or immunohistochemistry. RESULTS JHD ameliorated Lieber-DeCarli liquid diet-induced hepatic steatosis, inflammation and LPS expression. It improved pathological changes in the liver and alleviated intestinal ultrastructure injury. Moreover, it significantly enhanced the integrity of tight junctions by increasing the expression of zonula occludens-1 (ZO-1) and occludin. It suppressed the activation of myosin light chain (MLC) phosphorylation. CONCLUSION JHD improves intestinal barrier function and reduces gut leakiness in ALD rats.
Collapse
Affiliation(s)
- Xin Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong-Sheng Yao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lin Xu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong-Ming Yan
- Clinical Pharmacokinetics Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu Zhao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing-Hua Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qi-Lin Fu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yi-Yang Hu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Clinical Pharmacokinetics Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
23
|
Jiang Y, Liao Y, Liu Z, Zhou M, Wang H, Qi H, Sun S, Xi S, Tang Y. The effects of Cordyceps polysaccharides on ischemic brain injury in rats via intervening with IL-23/IL-17 axis and the intestinal barrier. Int J Biol Macromol 2024; 283:137526. [PMID: 39537075 DOI: 10.1016/j.ijbiomac.2024.137526] [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: 03/15/2024] [Revised: 09/25/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Cordyceps polysaccharide (CSP) has been shown to exhibit anti-inflammatory and antioxidant effects, with potential applications in ischemic stroke. This work is to explore the interventional potential of CSP in MCAO rats and the effects on the intestinal and cerebral IL-23/IL-17 axis. We conducted pharmacological experiments and mechanism exploration in MCAO rats. Our research showed that CSP improved the neurological function and cerebral pathological morphology, reduced cerebral infarction volume and water content in MCAO rats. We also found that CSP significantly decreased the IL-1β, TNF-α and IL-6 in the ischemic brain and enhanced the ability of MCAO rats to resist oxidative stress. Additionally, CSP improved intestinal barrier, inhibited the activation of the TLR4/Myd88/NF-κβ signaling pathway and IL-23/IL-17 axis. The study results demonstrated the effectiveness of CSP in interfering with MCAO rats. The mechanism appears to be related to protecting the intestinal barrier and inhibiting the IL-23/IL-17 axis.
Collapse
Affiliation(s)
- Yang Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yan Liao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhenquan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Manyu Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huizhang Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huiming Qi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shuyong Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Saiwen Xi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yibo Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
24
|
Lungaro L, Malfa P, Manza F, Costanzini A, Valentini G, Squarzanti DF, Viciani E, Velichevskaya A, Castagnetti A, Barbalinardo M, Gentili D, Cariani A, Ghisellini S, Caputo F, De Giorgio R, Caio G. Clinical Efficacy of Probiotics for Allergic Rhinitis: Results of an Exploratory Randomized Controlled Trial. Nutrients 2024; 16:4173. [PMID: 39683566 PMCID: PMC11644003 DOI: 10.3390/nu16234173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Allergic Rhinitis (AR) is an atopic disease affecting the upper airways of predisposed subjects exposed to aeroallergens. This study evaluates the effects of a mix of specific probiotics (L. acidophilus PBS066, L. rhamnosus LRH020, B. breve BB077, and B. longum subsp. longum BLG240) on symptoms and fecal microbiota modulation in subjects with AR. Methods: Probiotic effects were evaluated at the beginning (T0), at four and eight weeks of treatment (T1 and T2, respectively), and after four weeks of follow-up from the end of treatment (T3) (n = 19) compared to the placebo group (n = 22). AR symptoms and quality of life were evaluated by the mini rhinitis quality of life questionnaire (MiniRQLQ) at each time point. Allergic immune response and fecal microbiota compositions were assessed at T0, T2, and T3. The study was registered on Clinical-Trial.gov (NCT05344352). Results: The probiotic group showed significant improvement in the MiniRQLQ score at T1, T2, and T3 vs. T0 (p < 0.01, p < 0.05, p < 0.01, respectively). At T2, the probiotic group showed an increase in Dorea, which can be negatively associated with allergic diseases, and Fusicatenibacter, an intestinal bacterial genus with anti-inflammatory properties (p-value FDR-corrected = 0.0074 and 0.013, respectively). Conversely, at T3 the placebo group showed an increase in Bacteroides and Ruminococcaceae unassigned, (p-value FDR-corrected = 0.033 and 0.023, respectively) which can be associated with allergies, while the probiotic group showed a significative increase in the Prevotella/Bacteroides ratio (p-value FDR-corrected = 0.023). Conclusions: This probiotic formulation improves symptoms and quality of life in subjects with AR, promoting a shift towards anti-inflammatory and anti-allergic bacterial species in the intestinal microbiota.
Collapse
Affiliation(s)
- Lisa Lungaro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | | | - Francesca Manza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | - Anna Costanzini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | - Guido Valentini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | | | - Elisa Viciani
- Wellmicro Srl, 40138 Bologna, Italy; (E.V.); (A.V.); (A.C.)
| | | | | | - Marianna Barbalinardo
- Institute for the Study of Nanostructured Materials (CNR-ISMN), National Research Council, 40129 Bologna, Italy; (M.B.); (D.G.)
| | - Denis Gentili
- Institute for the Study of Nanostructured Materials (CNR-ISMN), National Research Council, 40129 Bologna, Italy; (M.B.); (D.G.)
| | - Alessio Cariani
- Analysis Laboratory, St. Anna Hospital, 44121 Ferrara, Italy; (A.C.); (S.G.)
| | - Sara Ghisellini
- Analysis Laboratory, St. Anna Hospital, 44121 Ferrara, Italy; (A.C.); (S.G.)
| | - Fabio Caputo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
- Complex Operational Unit of Internal Medicine, S.S. Annunziata Hospital, Cento, 44121 Ferrara, Italy
| | - Giacomo Caio
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital-Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
25
|
Zhao Z, Zhang X, Zhao F, Luo T. Microbiome-Metabolomics Analysis Insight into the Effects of Starvation and Refeeding on Intestinal Integrity in the Juvenile Largemouth Bass ( Micropterus salmoides). Int J Mol Sci 2024; 25:12500. [PMID: 39684211 DOI: 10.3390/ijms252312500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
The effects of starvation and refeeding on the gut condition of juvenile largemouth bass (Micropterus salmoides) remain unclear. Therefore, our research aimed to explore these effects. Amylase and lipase activities were remarkably decreased in the starvation (ST) group, yet prominently increased in the refeeding (RE) group (p < 0.05). In addition to the malondialdehyde (MDA) level, catalase (CAT) and superoxide dismutase (SOD) activities were significantly upregulated in the ST group (p < 0.05) in marked contrast to those in the controls; however, the RE group showed no substantial variations in CAT and SOD activities or the MDA level (p > 0.05). During starvation, the expression of Nrf2-Keap1 pathway-associated genes was significantly upregulated (p < 0.05). The comparative levels of TNF-α, IL-1β, and IL-15 were highly increased, with the levels of TGF-β1 and IL-10 apparently downregulated in the ST group; in contrast, these levels were restored to their original values in the RE group (p < 0.05). In contrast to the controls, the ST group showed significantly lower height and width of the villi, muscle thickness, and crypt depth and a higher goblet cell number; however, these values were recovered to some extent in the RE group (p < 0.05). The dominant bacterial phyla in the intestines of both groups were Proteobacteria, Firmicutes, Bacteroidetes, Acidobacteria, and Actinobacteria, with marked inter-group differences in the genera Serratia and Lactobacillus. Metabolomics analysis showed that amino acid metabolism is disrupted during starvation and is restored after refeeding. In summary, this study expands our comprehension of the interaction between oxidative stress and antioxidant defenses among juvenile largemouth bass subjected to starvation and refeeding.
Collapse
Affiliation(s)
- Zhenxin Zhao
- Institute of Fisheries, Guizhou Academy of Agricultural Sciences, Guiyang 550025, China
- Guizhou Special Aquatic Products Engineering Technology Center, Guiyang 550025, China
| | - Xianbo Zhang
- Institute of Fisheries, Guizhou Academy of Agricultural Sciences, Guiyang 550025, China
- Guizhou Special Aquatic Products Engineering Technology Center, Guiyang 550025, China
| | - Fei Zhao
- Institute of Fisheries, Guizhou Academy of Agricultural Sciences, Guiyang 550025, China
- Guizhou Special Aquatic Products Engineering Technology Center, Guiyang 550025, China
| | - Tianxun Luo
- Institute of Fisheries, Guizhou Academy of Agricultural Sciences, Guiyang 550025, China
- Guizhou Special Aquatic Products Engineering Technology Center, Guiyang 550025, China
| |
Collapse
|
26
|
Tong Y, Guo S, Li T, Yang K, Gao W, Peng F, Zou X. Gut microbiota and renal fibrosis. Life Sci 2024; 357:123072. [PMID: 39307181 DOI: 10.1016/j.lfs.2024.123072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Renal fibrosis represents a critical pathological condition in the progression of renal dysfunction, characterized by aberrant accumulation of extracellular matrix (ECM) and structural alterations in renal tissue. Recent research has highlighted the potential significance of gut microbiota and demonstrated their influence on host health and disease mechanisms through the production of bioactive metabolites. This review examines the role of alterations in gut microbial composition and their metabolites in the pathophysiological processes underlying renal fibrosis. It delineates current therapeutic interventions aimed at modulating gut microbiota composition, encompassing dietary modifications, pharmacological approaches, and probiotic supplementation, while evaluating their efficacy in mitigating renal fibrosis. Through a comprehensive analysis of current research findings, this review enhances our understanding of the bidirectional interaction between gut microbiota and renal fibrosis, establishing a theoretical foundation for future research directions and potential clinical applications in this domain.
Collapse
Affiliation(s)
- Yinghao Tong
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Shangze Guo
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Ting Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Kexin Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Fujun Peng
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
27
|
Boldyreva LV, Evtushenko AA, Lvova MN, Morozova KN, Kiseleva EV. Underneath the Gut-Brain Axis in IBD-Evidence of the Non-Obvious. Int J Mol Sci 2024; 25:12125. [PMID: 39596193 PMCID: PMC11594934 DOI: 10.3390/ijms252212125] [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: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
The gut-brain axis (GBA) plays a pivotal role in human health and wellness by orchestrating complex bidirectional regulation and influencing numerous critical processes within the body. Over the past decade, research has increasingly focused on the GBA in the context of inflammatory bowel disease (IBD). Beyond its well-documented effects on the GBA-enteric nervous system and vagus nerve dysregulation, and gut microbiota misbalance-IBD also leads to impairments in the metabolic and cellular functions: metabolic dysregulation, mitochondrial dysfunction, cationic transport, and cytoskeleton dysregulation. These systemic effects are currently underexplored in relation to the GBA; however, they are crucial for the nervous system cells' functioning. This review summarizes the studies on the particular mechanisms of metabolic dysregulation, mitochondrial dysfunction, cationic transport, and cytoskeleton impairments in IBD. Understanding the involvement of these processes in the GBA may help find new therapeutic targets and develop systemic approaches to improve the quality of life in IBD patients.
Collapse
Affiliation(s)
- Lidiya V. Boldyreva
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
| | - Anna A. Evtushenko
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
| | - Maria N. Lvova
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| | - Ksenia N. Morozova
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| | - Elena V. Kiseleva
- Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.N.L.); (K.N.M.); (E.V.K.)
| |
Collapse
|
28
|
Wickramasuriya SS, Park I, Lee Y, Richer LM, Przybyszewski C, Gay CG, van Oosterwijk JG, Lillehoj HS. Effect of orally administered B. subtilis-cNK-2 on growth performance, immunity, gut health, and gut microbiome in chickens infected with Eimeria acervulina and its potential as an alternative to antibiotics. Poult Sci 2024; 103:104156. [PMID: 39173215 PMCID: PMC11387274 DOI: 10.1016/j.psj.2024.104156] [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: 04/19/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
This study investigated the best oral delivery strategy (gavage or feed) for the B. subtilis expressing the chicken anti-microbial peptide cNK-2 (B. subtilis-cNK-2) in comparison to monensin, in chickens challenged with Eimeria acervulina (E. acervulina). A total of 120 broiler chickens were randomly allocated into 5 treatment groups in a completely randomized design: 1) uninfected chickens fed with basal diet (NC), 2) E. acervulina-infected chickens fed a basal diet (PC), 3) E. acervulina-infected chickens fed a basal diet supplemented with 90 mg monensin/kg feed (MO), 4) E. acervulina-infected chickens fed a basal diet and orally gavaged with B. subtilis-cNK-2 at 1 × 1010 cfu/d (CNK-O), and 5) E. acervulina-infected chickens fed a basal diet mixed with B. subtilis-cNK-2 at 1 × 1010 cfu/kg feed (CNK-F). The challenge consisted of 5,000 sporulated E. acervulina oocysts through oral gavage on d 15. Body weights were measured on d 7, 14, 21, and 23. Duodenal tissue and digesta samples were collected at 6 d postinfection (dpi) to assess the gut integrity, oxidative stress, mucosal immunity, and the gut microbiome. Fecal samples were collected from 6 to 8 dpi to enumerate the oocyst shedding. Chickens in the CNK-O group showed improved (P < 0.05) growth performance, gut integrity, and mucosal immunity compared to PC, comparable to chickens in the MO group. Chickens in the MO, CNK-F, and CNK-O treatment groups all showed lower (P < 0.05) oocyst shedding compared to PC chickens. Moreover, distinct cytokine profile, oxidative stress measures, tight junction proteins, and shifts in the gut microbiome with associated functional changes were observed in all challenge groups. In conclusion, we showed that the oral administration of B. subtilis-cNK-2 improved growth performance, enhanced local protective immunity, and reduced fecal oocyst shedding in broiler chickens infected with E. acervulina, demonstrating potential use of B. subtilis-cNK-2 as an alternative to antibiotics to protect chickens against coccidiosis.
Collapse
Affiliation(s)
- Samiru S Wickramasuriya
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA
| | - Inkyung Park
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA
| | - Youngsub Lee
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA
| | | | | | - Cyril G Gay
- Office of National Program-Animal Health, Agricultural Research Service, US Department of Agriculture, Beltsville, MD 20705, USA
| | | | - Hyun S Lillehoj
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD 20705, USA.
| |
Collapse
|
29
|
Kesharwani P, Halwai K, Jha SK, Al Mughram MH, Almujri SS, Almalki WH, Sahebkar A. Folate-engineered chitosan nanoparticles: next-generation anticancer nanocarriers. Mol Cancer 2024; 23:244. [PMID: 39482651 PMCID: PMC11526716 DOI: 10.1186/s12943-024-02163-z] [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: 05/03/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
Chitosan nanoparticles (NPs) are well-recognized as promising vehicles for delivering anticancer drugs due to their distinctive characteristics. They have the potential to enclose hydrophobic anticancer molecules, thereby enhancing their solubilities, permeabilities, and bioavailabilities; without the use of surfactant, i.e., through surfactant-free solubilization. This allows for higher drug concentrations at the tumor sites, prevents excessive toxicity imparted by surfactants, and could circumvent drug resistance. Moreover, biomedical engineers and formulation scientists can also fabricate chitosan NPs to slowly release anticancer agents. This keeps the drugs at the tumor site longer, makes therapy more effective, and lowers the frequency of dosing. Notably, some types of cancer cells (fallopian tube, epithelial tumors of the ovary, and primary peritoneum; lung, kidney, ependymal brain, uterus, breast, colon, and malignant pleural mesothelioma) have overexpression of folate receptors (FRs) on their outer surface, which lets folate-drug conjugate-incorporated NPs to target and kill them more effectively. Strikingly, there is evidence suggesting that the excessively produced FR&αgr (isoforms of the FR) stays consistent throughout treatment in ovarian and endometrial cancer, indicating resistance to conventional treatment; and in this regard, folate-anchored chitosan NPs can overcome it and improve the therapeutic outcomes. Interestingly, overly expressed FRs are present only in certain tumor types, which makes them a promising biomarker for predicting the effectiveness of FR-targeted therapy. On the other hand, the folate-modified chitosan NPs can also enhance the oral absorption of medicines, especially anticancer drugs, and pave the way for effective and long-term low-dose oral metronomic scheduling of poorly soluble and permeable drugs. In this review, we talked briefly about the techniques used to create, characterize, and tailor chitosan-based NPs; and delved deeper into the potential applications of folate-engineered chitosan NPs in treating various cancer types.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Kratika Halwai
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Uttar Pradesh, Kanpur, 208016, India
| | - Mohammed H Al Mughram
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Postal Code 61421, Abha, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Postal Code 61421, Abha, Saudi Arabia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Yu X, Pu H, Voss M. Overview of anti-inflammatory diets and their promising effects on non-communicable diseases. Br J Nutr 2024; 132:898-918. [PMID: 39411832 PMCID: PMC11576095 DOI: 10.1017/s0007114524001405] [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: 02/14/2024] [Revised: 06/26/2024] [Accepted: 08/03/2024] [Indexed: 11/20/2024]
Abstract
An anti-inflammatory diet is characterised by incorporating foods with potential anti-inflammatory properties, including fruits, vegetables, whole grains, nuts, legumes, spices, herbs and plant-based protein. Concurrently, pro-inflammatory red and processed meat, refined carbohydrates and saturated fats are limited. This article explores the effects of an anti-inflammatory diet on non-communicable diseases (NCD), concentrating on the underlying mechanisms that connect systemic chronic inflammation, dietary choices and disease outcomes. Chronic inflammation is a pivotal contributor to the initiation and progression of NCD. This review provides an overview of the intricate pathways through which chronic inflammation influences the pathogenesis of conditions including obesity, type II diabetes mellitus, CVD, autoinflammatory diseases, cancer and cognitive disorders. Through a comprehensive synthesis of existing research, we aim to identify some bioactive compounds present in foods deemed anti-inflammatory, explore their capacity to modulate inflammatory pathways and, consequently, to prevent or manage NCD. The findings demonstrated herein contribute to an understanding of the interplay between nutrition, inflammation and chronic diseases, paving a way for future dietary recommendations and research regarding preventive or therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoping Yu
- School of Medicine and Nursing, Chengdu University, Chengdu610106, People’s Republic of China
| | - Haomou Pu
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu611137, People’s Republic of China
| | - Margaret Voss
- Department of Nutrition and Food Studies, Falk College, Syracuse University, Syracuse, NY13244, USA
| |
Collapse
|
31
|
Zhu W, Cremonini E, Mastaloudis A, Oteiza PI. Glucoraphanin and sulforaphane mitigate TNFα-induced Caco-2 monolayers permeabilization and inflammation. Redox Biol 2024; 76:103359. [PMID: 39298837 PMCID: PMC11426148 DOI: 10.1016/j.redox.2024.103359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Intestinal permeabilization is central to the pathophysiology of chronic gut inflammation. This study investigated the efficacy of glucoraphanin (GR), prevalent in cruciferous vegetables, particularly broccoli, and its derivative sulforaphane (SF), in inhibiting tumor necrosis factor alpha (TNFα)-induced Caco-2 cell monolayers inflammation and permeabilization through the regulation of redox-sensitive events. TNFα binding to its receptor led to a rapid increase in oxidant production and subsequent elevation in the mRNA levels of NOX1, NOX4, and Duox2. GR and SF dose-dependently mitigated both these short- and long-term alterations in redox homeostasis. Downstream, GR and SF inhibited the activation of the redox-sensitive signaling cascades NF-κB (p65 and IKK) and MAPK ERK1/2, which contribute to inflammation and barrier permeabilization. GR (1 μM) and SF (0.5-1 μM) prevented TNFα-induced monolayer permeabilization and the associated reduction in the levels of the tight junction (TJ) proteins occludin and ZO-1. Both GR and SF also mitigated TNFα-induced increased mRNA levels of the myosin light chain kinase, which promotes TJ opening. Molecular docking suggests that although GR is mostly not absorbed, it could interact with extracellular and membrane sites in NOX1. Inhibition of NOX1 activity by GR would mitigate TNFα receptor downstream signaling and associated events. These findings support the concept that not only SF, but also GR, could exert systemic health benefits by protecting the intestinal barrier against inflammation-induced permeabilization, in part by regulating redox-sensitive pathways. GR has heretofore not been viewed as a biologically active molecule, but rather, the benign precursor of highly active SF. The consumption of GR and/or SF-rich vegetables or supplements in the diet may offer a means to mitigate the detrimental consequences of intestinal permeabilization, not only in disease states but also in conditions characterized by chronic inflammation of dietary and lifestyle origin.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Nutrition, University of California, Davis, CA, USA
| | | | | | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, USA; Department of Environmental Toxicology, University of California, Davis, CA, USA.
| |
Collapse
|
32
|
Yokoyama H, Masuyama T, Tanaka Y, Shimazaki T, Yasui Y, Abe T, Yoshinari K. Diacylglycerol O-acyltransferase 1 inhibitor increases plasma alanine aminotransferase and aspartate aminotransferase activities via a shedding of the intestinal villi and an increase in intestinal permeability in rats. Toxicol Lett 2024; 400:16-23. [PMID: 39096942 DOI: 10.1016/j.toxlet.2024.08.004] [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: 03/02/2024] [Revised: 06/18/2024] [Accepted: 08/01/2024] [Indexed: 08/05/2024]
Abstract
Diacylglycerol O-acyltransferase 1 (DGAT1) is a key enzyme for fat absorption step in the enterocytes. We previously reported that DGAT1 inhibition increased plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities in corn oil-loaded rats via protein kinase C (PKC) activation. In the present study, we investigated the mechanism with respect to the morphology and permeability of the small intestine, focusing on PKC function, and found that shortening of the intestinal villi and a decrease in the number of tdT-mediated dUTP-biotin nick-end labeling-positive cells in the tips of the villi were observed in the jejunum of DGAT1 inhibitor-treated rats loaded with corn oil. These results suggested that the tips of the villi were shed into the intestinal lumen. Next, fluorescein isothiocyanate-dextran, 110 kDa (FD-110) was administered intraduodenally to DGAT1 inhibitor-treated rats loaded with corn oil and we found that plasma FD-110 concentrations increased, indicating that the intestinal permeability to molecules with a molecular weight of approximately 110,000 (e.g., ALT and AST) increased. Taken together, the present results suggested that DGAT1 inhibitor-treatment in combination with corn oil causes ALT and AST to leak from the enterocytes into the blood by shedding the tips of the intestinal villi and increasing intestinal permeability.
Collapse
Affiliation(s)
- Hideaki Yokoyama
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC., 1-13-2 Fukuura, Kanazawa-ku, Yokohama-city, Kanagawa 236-0004, Japan; Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka-city, Shizuoka 422-8526, Japan.
| | - Taku Masuyama
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC., 1-13-2 Fukuura, Kanazawa-ku, Yokohama-city, Kanagawa 236-0004, Japan
| | - Yuki Tanaka
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC., 1-13-2 Fukuura, Kanazawa-ku, Yokohama-city, Kanagawa 236-0004, Japan
| | - Taishi Shimazaki
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC., 1-13-2 Fukuura, Kanazawa-ku, Yokohama-city, Kanagawa 236-0004, Japan
| | - Yuzo Yasui
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC., 1-13-2 Fukuura, Kanazawa-ku, Yokohama-city, Kanagawa 236-0004, Japan
| | - Takuya Abe
- Toxicology Research Lab., Central Pharmaceutical Research Institute, JAPAN TOBACCO INC., 1-13-2 Fukuura, Kanazawa-ku, Yokohama-city, Kanagawa 236-0004, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka-city, Shizuoka 422-8526, Japan
| |
Collapse
|
33
|
Morovati S, Baghkheirati AA, Sekhavati MH, Razmyar J. A Review on cLF36, a Novel Recombinant Antimicrobial Peptide-Derived Camel Lactoferrin. Probiotics Antimicrob Proteins 2024; 16:1886-1905. [PMID: 38722550 DOI: 10.1007/s12602-024-10285-5] [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] [Accepted: 05/01/2024] [Indexed: 10/02/2024]
Abstract
Lactoferrin is an antimicrobial peptide (AMP) playing a pivotal role in numerous biological processes. The primary antimicrobial efficacy of lactoferrin is associated with its N-terminal end, which contains various peptides, such as lactoferricin and lactoferrampin. In this context, our research team has developed a refined chimeric 42-mer peptide known as cLF36 over the past few years. This peptide encompasses the complete amino acid sequence of camel lactoferrampin and partial amino acid sequence of lactoferricin. The peptide's activity against human, avian, and plant bacterial pathogens has been assessed using different biological platforms, including prokaryotic (P170 and pET) and eukaryotic (HEK293) expression systems. The peptide positively influenced the growth performance and intestinal morphology of chickens challenged with pathogen bacteria. Computational methods and in vitro studies showed the peptide's antiviral effects against hepatitis C virus, influenza virus, and rotavirus. The chimeric peptide exhibited higher activity against certain tumor cell lines compared to normal cells, which may be attributed to the peptide's interaction with negatively charged glycosaminoglycans on the surface of tumor cells. Importantly, this peptide exhibited no toxicity against host cells and demonstrated remarkable thermal and protease stability in serum. In conclusion, while our investigations suggest that the chimeric peptide, cLF36, may offer potential as a candidate or complementary option to some available antibiotics, antiviral agents, and chemical pesticides, significant uncertainties remain regarding its cost-effectiveness, as well as its pharmacodynamic and pharmacokinetic characteristics, which require further elucidation.
Collapse
Affiliation(s)
- Solmaz Morovati
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | | | - Mohammad Hadi Sekhavati
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Jamshid Razmyar
- Department of Avian Diseases, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
34
|
Pradeloux S, Coulombe K, Ouamba AJK, Isenbrandt A, Calon F, Roy D, Soulet D. Oral Trehalose Intake Modulates the Microbiota-Gut-Brain Axis and Is Neuroprotective in a Synucleinopathy Mouse Model. Nutrients 2024; 16:3309. [PMID: 39408276 PMCID: PMC11478413 DOI: 10.3390/nu16193309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease affecting dopaminergic neurons in the nigrostriatal and gastrointestinal tracts, causing both motor and non-motor symptoms. This study examined the neuroprotective effects of trehalose. This sugar is confined in the gut due to the absence of transporters, so we hypothesized that trehalose might exert neuroprotective effects on PD through its action on the gut microbiota. We used a transgenic mouse model of PD (PrP-A53T G2-3) overexpressing human α-synuclein and developing GI dysfunctions. Mice were given water with trehalose, maltose, or sucrose (2% w/v) for 6.5 m. Trehalose administration prevented a reduction in tyrosine hydroxylase immunoreactivity in the substantia nigra (-25%), striatum (-38%), and gut (-18%) in PrP-A53T mice. It also modulated the gut microbiota, reducing the loss of diversity seen in PrP-A53T mice and promoting bacteria negatively correlated with PD in patients. Additionally, trehalose treatment increased the intestinal secretion of glucagon-like peptide 1 (GLP-1) by 29%. Maltose and sucrose, which break down into glucose, did not show neuroprotective effects, suggesting glucose is not involved in trehalose-mediated neuroprotection. Since trehalose is unlikely to cross the intestinal barrier at the given dose, the results suggest its effects are mediated indirectly through the gut microbiota and GLP-1.
Collapse
Affiliation(s)
- Solène Pradeloux
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Katherine Coulombe
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Alexandre Jules Kennang Ouamba
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté des Sciences de l’Agriculture et de l’Alimentation, Université Laval, Québec, QC G1V 0A6, Canada
| | - Amandine Isenbrandt
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Frédéric Calon
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Denis Roy
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté des Sciences de l’Agriculture et de l’Alimentation, Université Laval, Québec, QC G1V 0A6, Canada
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
35
|
Chang H, Perkins MH, Novaes LS, Qian F, Zhang T, Neckel PH, Scherer S, Ley RE, Han W, de Araujo IE. Stress-sensitive neural circuits change the gut microbiome via duodenal glands. Cell 2024; 187:5393-5412.e30. [PMID: 39121857 PMCID: PMC11425084 DOI: 10.1016/j.cell.2024.07.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/13/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Negative psychological states impact immunity by altering the gut microbiome. However, the relationship between brain states and microbiome composition remains unclear. We show that Brunner's glands in the duodenum couple stress-sensitive brain circuits to bacterial homeostasis. Brunner's glands mediated the enrichment of gut Lactobacillus species in response to vagus nerve stimulation. Cell-specific ablation of the glands markedly suppressed Lactobacilli counts and heightened vulnerability to infection. In the forebrain, we mapped a vagally mediated, polysynaptic circuit connecting the central nucleus of the amygdala to Brunner's glands. Chronic stress suppressed central amygdala activity and phenocopied the effects of gland lesions. Conversely, excitation of either the central amygdala or parasympathetic vagal neurons activated Brunner's glands and reversed the effects of stress on the gut microbiome and immunity. The findings revealed a tractable brain-body mechanism linking psychological states to host defense.
Collapse
Affiliation(s)
- Hao Chang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Matthew H Perkins
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Leonardo S Novaes
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Tong Zhang
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou 510180, Guangdong, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Peter H Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen 72074, Germany
| | - Simon Scherer
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Tübingen 72076, Germany
| | - Ruth E Ley
- Max-Planck Institute for Biology, Tübingen 72076, Germany
| | - Wenfei Han
- Max-Planck Institute for Biological Cybernetics, Tübingen 72076, Germany.
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Max-Planck Institute for Biological Cybernetics, Tübingen 72076, Germany.
| |
Collapse
|
36
|
Zhu W, Xiong L, Oteiza PI. Structure-dependent capacity of procyanidin dimers to inhibit inflammation-induced barrier dysfunction in a cell model of intestinal epithelium. Redox Biol 2024; 75:103275. [PMID: 39059205 PMCID: PMC11327484 DOI: 10.1016/j.redox.2024.103275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Diet is of major importance in modulating intestinal inflammation, as the gastrointestinal tract is directly exposed to high concentrations of dietary components. Procyanidins are flavan-3-ol oligomers abundant in fruits and vegetables. Although with limited or no intestinal absorption, they can have GI health benefits which can promote overall health. We previously observed that epicatechin gallate (ECG) and epigallocatechin gallate (EGCG) dimers inhibit in vitro colorectal cancer cell proliferation and invasiveness. Inflammation-mediated intestinal barrier permeabilization can result in a chronic inflammatory condition and promote colorectal cancer onset/progression. Thus, this study investigated the structure-dependent capacity of ECG, EGCG and (-)-epicatechin (EC) dimers to inhibit tumor necrosis factor alpha (TNFα)-induced inflammation, oxidative stress, and loss of barrier integrity in Caco-2 cells differentiated into an intestinal epithelial cell monolayer. Cells were incubated with TNFα (10 ng/ml), in the absence/presence of ECG, EGCG and EC dimers. The three dimers inhibited TNFα-mediated Caco-2 cell monolayer permeabilization, modulating events involved in the loss of barrier function and inflammation, i.e. decreased tight junction protein levels; increased matrix metalloproteinases expression and activity; increased NADPH oxidase expression and oxidant production; activation of the NF-κB and ERK1/2 pathways and downstream events leading to tight junction opening. For some of these mechanisms, the galloylated ECG and EGCG dimers had stronger protective potency than the non-galloylated EC dimer. These differences could be due to differential membrane interactions as pointed out by molecular dynamics simulation of procyanidin dimers-cell membrane interactions and/or by differential interactions with NOX1. Results show that dimeric procyanidins, although poorly absorbed, can promote health by alleviating intestinal inflammation, oxidative stress and barrier permeabilization.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Nutrition, University of California, Davis, CA, 95618, USA
| | - Le Xiong
- Cleveland Clinic, Cleveland, OH, 44194, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, 95618, USA; Department of Environmental Toxicology, University of California, Davis, CA, 95618, USA.
| |
Collapse
|
37
|
Cao S, Pan Y, Zheng W, Chen S, Yin T, Liu R, You J. Effect of ozone oxidation on gastrointestinal digestion and absorption characteristics of silver carp (Hypophthalmichthys molitrix) surimi gels in vitro. Food Res Int 2024; 192:114759. [PMID: 39147534 DOI: 10.1016/j.foodres.2024.114759] [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: 05/24/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 08/17/2024]
Abstract
To investigate the quality of different ozone-oxidized surimi gels and their in vitro digestion and absorption characteristics, surimi rinsed with different concentrations of ozonated water (0, 8, 26 mg/L) were prepared. Then, the degree of oxidation and gel structure of surimi were determined, the in vitro digestion and absorption of the gels were simulated, and the digestion and absorption products were analyzed by LC-MS/MS. The results showed that the quality of surimi gels was improved after proper ozone oxidation. After ozone water rinsing, the dry matter digestibility, peptide, and amino acid content increased, and the changes of all three were in line with the Logistic kinetic model (R2 = 0.95-0.99). Caco-2 cell absorption experiments showed that the absorption rate of peptides and amino acids decreased after ozone water rinsing. In summary, ozone oxidation can promote the digestion of surimi gels, but it also reduces the absorption of peptides and amino acids by Caco-2 cells. This study provides a reference for the application of ozone in the food field.
Collapse
Affiliation(s)
- Shuning Cao
- College of Food Science and Technology, Huazhong Agricultural University/Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei Province 430070, China.
| | - Yuping Pan
- College of Food Science and Technology, Huazhong Agricultural University/Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei Province 430070, China.
| | - Wendong Zheng
- College of Food Science and Technology, Huazhong Agricultural University/Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei Province 430070, China.
| | - Shengjun Chen
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510300, China.
| | - Tao Yin
- College of Food Science and Technology, Huazhong Agricultural University/Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei Province 430070, China; National R & D Branch Center for Conventional Freshwater Fish Processing, Wuhan, Hubei 430070, China.
| | - Ru Liu
- College of Food Science and Technology, Huazhong Agricultural University/Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei Province 430070, China; National R & D Branch Center for Conventional Freshwater Fish Processing, Wuhan, Hubei 430070, China.
| | - Juan You
- College of Food Science and Technology, Huazhong Agricultural University/Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei Province 430070, China; National R & D Branch Center for Conventional Freshwater Fish Processing, Wuhan, Hubei 430070, China.
| |
Collapse
|
38
|
Abdulqadir R, Al-Sadi R, Haque M, Gupta Y, Rawat M, Ma TY. Bifidobacterium bifidum Strain BB1 Inhibits Tumor Necrosis Factor-α-Induced Increase in Intestinal Epithelial Tight Junction Permeability via Toll-Like Receptor-2/Toll-Like Receptor-6 Receptor Complex-Dependent Stimulation of Peroxisome Proliferator-Activated Receptor γ and Suppression of NF-κB p65. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1664-1683. [PMID: 38885924 PMCID: PMC11372998 DOI: 10.1016/j.ajpath.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Bifidobacterium bifidum strain BB1 causes a strain-specific enhancement in intestinal epithelial tight junction (TJ) barrier. Tumor necrosis factor (TNF)-α induces an increase in intestinal epithelial TJ permeability and promotes intestinal inflammation. The major purpose of this study was to delineate the protective effect of BB1 against the TNF-α-induced increase in intestinal TJ permeability and to unravel the intracellular mechanisms involved. TNF-α produces an increase in intestinal epithelial TJ permeability in Caco-2 monolayers and in mice. Herein, the addition of BB1 inhibited the TNF-α increase in Caco-2 intestinal TJ permeability and mouse intestinal permeability in a strain-specific manner. BB1 inhibited the TNF-α-induced increase in intestinal TJ permeability by interfering with TNF-α-induced enterocyte NF-κB p50/p65 and myosin light chain kinase (MLCK) gene activation. The BB1 protective effect against the TNF-α-induced increase in intestinal permeability was mediated by toll-like receptor-2/toll-like receptor-6 heterodimer complex activation of peroxisome proliferator-activated receptor γ (PPAR-γ) and PPAR-γ pathway inhibition of TNF-α-induced inhibitory kappa B kinase α (IKK-α) activation, which, in turn, resulted in a step-wise inhibition of NF-κB p50/p65, MLCK gene, MLCK kinase activity, and MLCK-induced opening of the TJ barrier. In conclusion, these studies unraveled novel intracellular mechanisms of BB1 protection against the TNF-α-induced increase in intestinal TJ permeability. The current data show that BB1 protects against the TNF-α-induced increase in intestinal epithelial TJ permeability via a PPAR-γ-dependent inhibition of NF-κB p50/p65 and MLCK gene activation.
Collapse
Affiliation(s)
- Raz Abdulqadir
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania.
| | - Rana Al-Sadi
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Mohammad Haque
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Yash Gupta
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Manmeet Rawat
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Thomas Y Ma
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania.
| |
Collapse
|
39
|
Sharma MK, Lee J, Shi H, Ko H, Goo D, Paneru D, Holladay SD, Gogal RM, Kim WK. Effect of dietary inclusion of 25-hydroxyvitamin D₃ and vitamin E on performance, gut health, oxidative status, and immune response in laying hens infected with coccidiosis. Poult Sci 2024; 103:104033. [PMID: 39059054 PMCID: PMC11331952 DOI: 10.1016/j.psj.2024.104033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Vitamin D3 (25-hydroxyvitamin D3 (VD)) and vitamin E (VE) have proven to have immunomodulatory and antioxidant functions along with capacities to improve the reproductive function in chickens. Coccidiosis in laying hens at different stages of growth has been shown to negatively affect performance, immune response, and oxidative status, thus increasing the cost of production. A study was conducted to evaluate the influence of dietary VD or VE on performance, gut health, immune response, and oxidative status of laying hens at peak production. Laying hens (23 wk-of-age, n = 225) were randomly allocated into 5 treatment groups (n = 9 hens/replicate) with 5 replicate groups each: 1) unchallenged control (UC), 2) pair-fed control (PF), 3) challenged control (CC), 4) challenged control top-dressed with 5,000 IU of 25-hydroxyvitamin D3 (VD) per kg of diet, and 5) challenged control top-dressed with 100 IU of DL-α-tocopherol (VE). At 25 wk-of-age, hens grouped in CC, VD, and VE were challenged with mixed Eimeria spp. to induce coccidiosis. VD or VE supplemented hens did not impact bird body weight; however, egg production increased by 10.36% and 13.77%, respectively (P < 0.0001). Furthermore, the gut health of the hens was improved with either VD or VE supplementation, as indicated by lowered gut permeability and intestinal lesion scores (P < 0.05). VE significantly reduced the heterophil count (P = 0.0490) alongside numerically increasing the peripheral CD4+ and CD8+ T cells and monocyte counts (P > 0.05). Both VD or VE increased the TAC at 14 DPI compared to UC (P<0.05). Preliminary findings suggest that dietary VD or VE supplementation has the potential to improve gut health, modulate the immune response, and increase egg production in coccidiosis-infected laying hens.
Collapse
Affiliation(s)
- Milan Kumar Sharma
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Jihwan Lee
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Hanyi Shi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Hanseo Ko
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Doyun Goo
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Deependra Paneru
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Steven D Holladay
- Department of Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Robert M Gogal
- Department of Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
40
|
Zhang J, Zhang R, Chen Y, Guo X, Ren Y, Wang M, Li X, Huang Z, Zhu W, Yu K. Indole-3-aldehyde Alleviates High-Fat Diet-Induced Gut Barrier Disruption by Increasing Intestinal Stem Cell Expansion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18930-18941. [PMID: 39146439 DOI: 10.1021/acs.jafc.4c02381] [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: 08/17/2024]
Abstract
High-fat diet (HFD) feeding is known to cause intestinal barrier disruption, thereby triggering severe intestinal inflammatory disease. Indole-3-aldehyde (IAld) has emerged as a potential candidate for mitigating inflammatory responses and maintaining intestinal homeostasis. However, the role of IAld in the HFD-related intestinal disruption remains unclear. In this study, 48 7 week-old male C57BL/6J mice were assigned to four groups: the normal chow diet (NCD) group received a NCD; the HFD group was fed an HFD; the HFD + IAld200 group was supplemented with 200 mg/kg IAld in the HFD; and the HFD + IAld600 group was supplemented with 600 mg/kg IAld in the HFD. The results showed that dietary IAld supplementation ameliorated fat accumulation and metabolic disorders, which are associated with reduced intestinal permeability. This reduction potentially led to decreased systemic inflammation and enhanced intestinal barrier function in HFD-fed mice. Furthermore, we found that IAld promoted intestinal stem cell (ISC) proliferation by activating aryl hydrocarbon receptors (AHRs) in vivo and ex vivo. These findings suggest that IAld restores the HFD-induced intestinal barrier disruption by promoting AHR-mediated ISC proliferation.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruofan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
- Animal Health Inspection Institute of Suzhou, Wujiang District, Suzhou 215200, China
| | - Yahui Chen
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xin Guo
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuting Ren
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Mengting Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Zan Huang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
41
|
Choi Y, Park H, Kim J, Lee H, Kim M. Heat Stress Induces Alterations in Gene Expression of Actin Cytoskeleton and Filament of Cellular Components Causing Gut Disruption in Growing-Finishing Pigs. Animals (Basel) 2024; 14:2476. [PMID: 39272260 PMCID: PMC11394201 DOI: 10.3390/ani14172476] [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: 07/03/2024] [Revised: 08/17/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
We aimed to investigate the impact of heat stress (HS) on the expression of tight junction (TJ) proteins and the interaction between genes affecting intestinal barrier function using transcriptomics in the porcine jejunum. Twenty-four barrows (crossbred Yorkshire × Landrace × Duroc; average initial body weight, 56.71 ± 1.74 kg) were placed in different temperatures (normal temperature [NT]; HS) and reared for 56 days. At the end of the experiment, jejunal samples were collected from three pigs per treatment for transcriptome and reverse-transcription quantitative polymerase chain reaction (RT-qPCR) analyses. We identified 43 differentially expressed genes, involving five Kyoto Encyclopedia of Genes and Genomes pathways, eight molecular functions, seven cellular components (CCs), and nine biological processes, using gene ontology enrichment analysis. Genes associated with the actin cytoskeleton, filament-binding pathways, and TJ proteins were selected and analyzed by RT-qPCR. Significant differences in relative mRNA expression showed that downregulated genes in the HS group included ZO1, CLDN1, OCLN, PCK1, and PCK2, whereas ACTG2, DES, MYL9, MYLK, TPM1, TPM2, CNN1, PDLIM3, and PCP4 were upregulated by HS (p < 0.05). These findings indicate that HS in growing-finishing pigs induces depression in gut integrity, which may be related to genes involved in the actin cytoskeleton and filaments of CC.
Collapse
Affiliation(s)
- Yohan Choi
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Hyunju Park
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Joeun Kim
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Hyunseo Lee
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Republic of Korea
| | - Minju Kim
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Republic of Korea
- Institute of Applied Humanimal Science, Hankyong National University, Anseong 17579, Republic of Korea
| |
Collapse
|
42
|
Li Z, Wan M, Wang M, Duan J, Jiang S. Modulation of gut microbiota on intestinal permeability: A novel strategy for treating gastrointestinal related diseases. Int Immunopharmacol 2024; 137:112416. [PMID: 38852521 DOI: 10.1016/j.intimp.2024.112416] [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: 02/08/2024] [Revised: 05/07/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Accumulating evidence emphasizes the critical reciprocity between gut microbiota and intestinal barrier function in maintaining the gastrointestinal homeostasis. Given the fundamental role caused by intestinal permeability, which has been scrutinized as a measurable potential indicator of perturbed barrier function in clinical researches, it seems not surprising that recent decades have been marked by augmented efforts to determine the interaction between intestinal microbes and permeability of the individual. However, despite the significant progress in characterizing intestinal permeability and the commensal bacteria in the intestine, the mechanisms involved are still far from being thoroughly revealed. In the present review, based on multiomic methods, high-throughput sequencing and molecular biology techniques, the impacts of gut microbiota on intestinal permeability as well as their complex interaction networks are systematically summarized. Furthermore, the diseases related to intestinal permeability and main causes of changes in intestinal permeability are briefly introduced. The purpose of this review is to provide a novel prospection to elucidate the correlation between intestinal microbiota and permeability, and to explore a promising solution for diagnosis and treatment of gastrointestinal related diseases.
Collapse
Affiliation(s)
- Zhuotong Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Meiyu Wan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Mingyang Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| |
Collapse
|
43
|
Perera R, Alharbi K, Hasan A, Asnayanti A, Do A, Shwani A, Murugesan R, Ramirez S, Kidd M, Alrubaye AAK. Evaluating the Impact of the PoultryStar ®Bro Probiotic on the Incidence of Bacterial Chondronecrosis with Osteomyelitis Using the Aerosol Transmission Challenge Model. Microorganisms 2024; 12:1630. [PMID: 39203472 PMCID: PMC11356867 DOI: 10.3390/microorganisms12081630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Bacterial chondronecrosis with osteomyelitis (BCO) lameness is a major welfare issue for broiler production worldwide affecting approximately 1.5% of broilers over 42 days old. Excessive body weight gain causes mechanical stress on long bones, leading to micro-fractures. This condition induces a bacterial infection of fractures, resulting in bone necrosis and eventual BCO lameness. Increasing gut integrity and supporting Calcium metabolism contribute to the optimal bone structure and subsequently reduce BCO lameness. Probiotics thus provide an excellent strategy for alleviating BCO due to the improvement of intestinal integrity and barrier function. Accordingly, the present study investigated the lameness reduction through the feed supplementation of a selected probiotic. Broiler chickens were assigned to three treatments, including a control litter group (FL), a PoultryStar®Bro probiotic fed group (BRO), and a control wire-flooring group (CW) designed to induce BCO lameness. The probiotic significantly decreased lameness by 46% compared to the control group (p < 0.05). The most predominant bacteria identified from the BCO lesions were Staphylococcus cohnii and Staphylococcus lentus. Moreover, significant increments of tight junction gene expression in jejunum and ileum, plus numerical improvements of body weight gain (BW; +360 g) and feed conversion ratio (FCR; -12 pts) were observed in BRO-supplemented birds.
Collapse
Affiliation(s)
- Ruvindu Perera
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (R.P.); (K.A.); (A.A.); (A.D.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Khawla Alharbi
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (R.P.); (K.A.); (A.A.); (A.D.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Amer Hasan
- Department of Veterinary Public Health, College of Veterinary Medicine, University of Baghdad, Baghdad P.O. Box 1417, Iraq;
| | - Andi Asnayanti
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (R.P.); (K.A.); (A.A.); (A.D.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Anh Do
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (R.P.); (K.A.); (A.A.); (A.D.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Abdulkarim Shwani
- Exotic and Emerging Avian Viral Disease Research Unit, Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, Agricultural Research Service, USDA, Athens, GA 30605, USA;
| | - Raj Murugesan
- BIOMIN America Inc., 10801 Mastin Blvd Suite 100, Overland Park, KS 66210, USA; (R.M.); (S.R.)
| | - Shelby Ramirez
- BIOMIN America Inc., 10801 Mastin Blvd Suite 100, Overland Park, KS 66210, USA; (R.M.); (S.R.)
| | - Michael Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Adnan A. K. Alrubaye
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (R.P.); (K.A.); (A.A.); (A.D.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| |
Collapse
|
44
|
Bernardi C, Cappellucci G, Baini G, Aloisi AM, Finetti F, Trabalzini L. Potential Human Health Benefits of Phaseolus vulgaris L. var Venanzio: Effects on Cancer Cell Growth and Inflammation. Nutrients 2024; 16:2534. [PMID: 39125413 PMCID: PMC11314099 DOI: 10.3390/nu16152534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/10/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
It is widely recognized that foods, biodiversity, and human health are strongly interconnected, and many efforts have been made to understand the nutraceutical value of diet. In particular, diet can affect the progression of intestinal diseases, including inflammatory bowel disease (IBD) and intestinal cancer. In this context, we studied the anti-inflammatory and antioxidant activities of extracts obtained from a local endangered variety of Phaseolus vulgaris L. (Fagiola di Venanzio, FV). Using in vitro intestinal cell models, we evaluated the activity of three different extracts: soaking water, cooking water, and the bioaccessible fraction obtained after mimicking the traditional cooking procedure and gastrointestinal digestion. We demonstrated that FV extracts reduce inflammation and oxidative stress prompted by interleukin 1β through the inhibition of cyclooxygenase 2 expression and prostaglandin E2 production and through the reduction in reactive oxygen species production and NOX1 levels. The reported data outline the importance of diet in the prevention of human inflammatory diseases. Moreover, they strongly support the necessity to safeguard local biodiversity as a source of bioactive compounds.
Collapse
Affiliation(s)
- Clizia Bernardi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (L.T.)
| | - Giorgio Cappellucci
- Department of Physical Sciences, Earth and Environment, University of Siena, Via Laterina 8, 53100 Siena, Italy; (G.C.); (G.B.)
| | - Giulia Baini
- Department of Physical Sciences, Earth and Environment, University of Siena, Via Laterina 8, 53100 Siena, Italy; (G.C.); (G.B.)
| | - Anna Maria Aloisi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy;
| | - Federica Finetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (L.T.)
| | - Lorenza Trabalzini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (L.T.)
| |
Collapse
|
45
|
Singh S, Koo OK. A Comprehensive Review Exploring the Protective Role of Specific Commensal Gut Bacteria against Salmonella. Pathogens 2024; 13:642. [PMID: 39204243 PMCID: PMC11356920 DOI: 10.3390/pathogens13080642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Gut microbiota is a diverse community of microorganisms that constantly work to protect the gut against pathogens. Salmonella stands out as a notorious foodborne pathogen that interacts with gut microbes, causing an imbalance in the overall composition of microbiota and leading to dysbiosis. This review focuses on the interactions between Salmonella and the key commensal bacteria such as E. coli, Lactobacillus, Clostridium, Akkermansia, and Bacteroides. The review highlights the role of these gut bacteria and their synergy in combating Salmonella through several mechanistic interactions. These include the production of siderophores, which compete with Salmonella for essential iron; the synthesis of short-chain fatty acids (SCFAs), which exert antimicrobial effects and modulate the gut environment; the secretion of bacteriocins, which directly inhibit Salmonella growth; and the modulation of cytokine responses, which influences the host's immune reaction to infection. While much research has explored Salmonella, this review aims to better understand how specific gut bacteria engage with the pathogen, revealing distinct defense mechanisms tailored to each species and how their synergy may lead to enhanced protection against Salmonella. Furthermore, the combination of these commensal bacteria could offer promising avenues for bacteria-mediated therapy during Salmonella-induced gut infections in the future.
Collapse
Affiliation(s)
| | - Ok Kyung Koo
- Department of Food Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea;
| |
Collapse
|
46
|
Feng R, Wang Q, Yu T, Hu H, Wu G, Duan X, Jiang R, Xu Y, Huang Y. Quercetin ameliorates bone loss in OVX rats by modulating the intestinal flora-SCFAs-inflammatory signaling axis. Int Immunopharmacol 2024; 136:112341. [PMID: 38810309 DOI: 10.1016/j.intimp.2024.112341] [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: 04/30/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Osteoporosis (OP) is a common systemic skeletal disorder characterized by an imbalance in bone homeostasis, involving increased osteoclastic bone formation and decreased osteoblastic bone resorption. Quercetin is a plant polyphenol that has been found to exhibit various biological activities, including antioxidant, anti-inflammatory, and antimicrobial effects. Previous studies have demonstrated its potential to improve postmenopausal OP, although the exact mechanism remains unclear. This study aims to investigate the anti-osteoporotic mechanism of quercetin based on the "intestinal flora - short-chain fatty acids (SCFAs) - inflammatory" signaling axis. METHODS In this study, we established an ovariectomized (OVX)-induced rat model, quercetin intervention and evaluated the effects on rats following antibiotic (ABX) treatment and fecal microbiota transplantation (FMT). After 6 weeks of intervention, the rats were euthanized, and samples from their femur, tibia, lumbar spine, serum, colon and feces were collected, and bone strength, intestinal flora structure, SCFAs levels and cytokine levels were assessed. RESULTS Quercetin modulates the intestinal flora by increasing potentially probiotic bacteria (i.e., Lactobacillales, Prevotellaceae, and Blautia) and decreasing potentially pathogenic bacteria (Desulfobacterota, Erysipelotrichales, Romboutsia, and Butyricoccaceae). It also increases SCFAs content and reduces colonic permeability by enhancing tight junction proteins (ZO-1, Occludin). Furthermore, quercetin lowers proinflammatory cytokine levels (LPS, IL-1β, and TNF-α), which enhances bone strength and prevents OVX-induced bone loss. CONCLUSIONS Quercetin may effectively reduce bone loss in OVX rats via the "intestinal flora - SCFAs - inflammatory" signaling pathway.
Collapse
Affiliation(s)
- Ruibing Feng
- Department of Spine Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei Province 430074, PR China
| | - Qing Wang
- School of Sports Medicine, Wuhan Institute of Physical Education, Wuhan City, Hubei Province 430079, PR China
| | - Tiantian Yu
- Hubei University of Traditional Chinese Medicine, Wuhan, Hubei Province 430060, PR China
| | - Hao Hu
- Department of Spine Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei Province 430074, PR China; School of Sports Medicine, Wuhan Institute of Physical Education, Wuhan City, Hubei Province 430079, PR China; Hubei University of Traditional Chinese Medicine, Wuhan, Hubei Province 430060, PR China
| | - Gang Wu
- Department of Spine Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei Province 430074, PR China; School of Sports Medicine, Wuhan Institute of Physical Education, Wuhan City, Hubei Province 430079, PR China; Hubei University of Traditional Chinese Medicine, Wuhan, Hubei Province 430060, PR China
| | - Xiaofeng Duan
- Department of Spine Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei Province 430074, PR China
| | - Ruixuan Jiang
- Hubei University of Traditional Chinese Medicine, Wuhan, Hubei Province 430060, PR China
| | - Yifan Xu
- School of Sports Medicine, Wuhan Institute of Physical Education, Wuhan City, Hubei Province 430079, PR China
| | - Yong Huang
- Department of Spine Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei Province 430074, PR China; School of Sports Medicine, Wuhan Institute of Physical Education, Wuhan City, Hubei Province 430079, PR China; Hubei University of Traditional Chinese Medicine, Wuhan, Hubei Province 430060, PR China.
| |
Collapse
|
47
|
D’Ambrosio A, Altomare A, Boscarino T, Gori M, Balestrieri P, Putignani L, Del Chierico F, Carotti S, Cicala M, Guarino MPL, Piemonte V. Mathematical Modeling of Vedolizumab Treatment's Effect on Microbiota and Intestinal Permeability in Inflammatory Bowel Disease Patients. Bioengineering (Basel) 2024; 11:710. [PMID: 39061792 PMCID: PMC11274165 DOI: 10.3390/bioengineering11070710] [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: 06/01/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Growing evidence suggests that impaired gut permeability and gut microbiota alterations are involved in the pathogenesis of Inflammatory Bowel Diseases (IBDs), which include Ulcerative Colitis (UC) and Crohn's Disease (CD). Vedolizumab is an anti-α4β7 antibody approved for IBD treatment, used as the first treatment or second-line therapy when the first line results in inadequate effectiveness. The aim of this study is to develop a mathematical model capable of describing the pathophysiological mechanisms of Vedolizumab treatment in IBD patients. In particular, the relationship between drug concentration in the blood, colonic mucosal permeability and fecal microbiota composition was investigated and modeled to detect and predict trends in order to support and tailor Vedolizumab therapies. To pursue this aim, clinical data from a pilot study on a cluster of 11 IBD patients were analyzed. Enrolled patients underwent colonoscopy in three phases (before (t0), after 24 weeks of (t1) and after 52 weeks of (t2 ) Vedolizumab treatment) to collect mucosal biopsies for transepithelial electrical resistance (TEER) evaluation (permeability to ions), intestinal permeability measurement and histological analysis. Moreover, fecal samples were collected for the intestinal microbiota analysis at the three time points. The collected data were compared to those of 11 healthy subjects at t0, who underwent colonoscopy for screening surveillance, and used to implement a three-compartmental mathematical model (comprising central blood, peripheral blood and the intestine). The latter extends previous evidence from the literature, based on the regression of experimental data, to link drug concentration in the peripheral blood compartment with Roseburia abundance and intestinal permeability. The clinical data showed that Vedolizumab treatment leads to an increase in TEER and a reduction in intestinal permeability to a paracellular probe, improving tissue inflammation status. Microbiota analysis showed increasing values of Roseburia, albeit not statistically significant. This trend was adequately reproduced by the mathematical model, which offers a useful tool to describe the pathophysiological effects of Vedolizumab therapy on colonic mucosal permeability and fecal microbiota composition. The model's satisfactory predictive capabilities and simplicity shed light on the relationship between the drug, the microbiota and permeability and allow for its straightforward extension to diverse therapeutic conditions.
Collapse
Affiliation(s)
- Antonio D’Ambrosio
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (T.B.); (V.P.)
| | - Annamaria Altomare
- Department of Sciences and Technology of Sustainable Development and Human Health, Università Campus Biomedico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy;
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
| | - Tamara Boscarino
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (T.B.); (V.P.)
| | - Manuele Gori
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), International Campus “A. Buzzati-Traverso”, Via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Paola Balestrieri
- Gastroenterology Unit, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy;
| | - Lorenza Putignani
- Units of Microbiomics and Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Federica Del Chierico
- Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Simone Carotti
- Microscopic and Ultrastructural Anatomy Research Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
| | - Michele Cicala
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
- Microscopic and Ultrastructural Anatomy Research Unit, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
| | - Michele Pier Luca Guarino
- Gastroenterology Research Unit, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.G.); (M.C.); (M.P.L.G.)
- Gastroenterology Unit, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy;
| | - Vincenzo Piemonte
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (T.B.); (V.P.)
| |
Collapse
|
48
|
Dmytriv TR, Storey KB, Lushchak VI. Intestinal barrier permeability: the influence of gut microbiota, nutrition, and exercise. Front Physiol 2024; 15:1380713. [PMID: 39040079 PMCID: PMC11260943 DOI: 10.3389/fphys.2024.1380713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
The intestinal wall is a selectively permeable barrier between the content of the intestinal lumen and the internal environment of the body. Disturbances of intestinal wall permeability can potentially lead to unwanted activation of the enteric immune system due to excessive contact with gut microbiota and its components, and the development of endotoxemia, when the level of bacterial lipopolysaccharides increases in the blood, causing chronic low-intensity inflammation. In this review, the following aspects are covered: the structure of the intestinal wall barrier; the influence of the gut microbiota on the permeability of the intestinal wall via the regulation of functioning of tight junction proteins, synthesis/degradation of mucus and antioxidant effects; the molecular mechanisms of activation of the pro-inflammatory response caused by bacterial invasion through the TLR4-induced TIRAP/MyD88 and TRAM/TRIF signaling cascades; the influence of nutrition on intestinal permeability, and the influence of exercise with an emphasis on exercise-induced heat stress and hypoxia. Overall, this review provides some insight into how to prevent excessive intestinal barrier permeability and the associated inflammatory processes involved in many if not most pathologies. Some diets and physical exercise are supposed to be non-pharmacological approaches to maintain the integrity of intestinal barrier function and provide its efficient operation. However, at an early age, the increased intestinal permeability has a hormetic effect and contributes to the development of the immune system.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
49
|
Yuan S, Sun M, Ma D, Guo X, Wang Z, Niu J, Jiang W, He Y, Wei M, Qi J. Exploring the underlying mechanisms of enteritis impact on golden pompano (Trachinotus ovatus) through multi-omics analysis. FISH & SHELLFISH IMMUNOLOGY 2024; 150:109616. [PMID: 38734118 DOI: 10.1016/j.fsi.2024.109616] [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: 02/11/2024] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Enteritis posed a significant health challenge to golden pompano (Trachinotus ovatus) populations. In this research, a comprehensive multi-omics strategy was implemented to elucidate the pathogenesis of enteritis by comparing both healthy and affected golden pompano. Histologically, enteritis was characterized by villi adhesion and increased clustering after inflammation. Analysis of the intestinal microbiota revealed a significant increase (P < 0.05) in the abundance of specific bacterial strains, including Photobacterium and Salinivibrio, in diseased fish compared to the healthy group. Metabolomic analysis identified 5479 altered metabolites, with significant impacts on terpenoid and polyketide metabolism, as well as lipid metabolism (P < 0.05). Additionally, the concentrations of several compounds such as calcitetrol, vitamin D2, arachidonic acid, and linoleic acid were significantly reduced in the intestines of diseased fish post-enteritis (P < 0.05), with the detection of harmful substances such as Efonidipine. In transcriptomic profiling, enteritis induced 68 upregulated and 73 downregulated genes, predominantly affecting steroid hormone receptor activity (P < 0.05). KEGG pathway enrichment analysis highlighted upregulation of SQLE and CYP51 in steroidogenesis, while the HSV-1 associated MHC1 gene exhibited significant downregulation. Integration of multi-omics results suggested a potential pathogenic mechanism: enteritis may have resulted from concurrent infection of harmful bacteria, specifically Photobacterium and Salinivibrio, along with HSV-1. Efonidipine production within the intestinal tract may have blocked certain calcium ion channels, leading to downregulation of MHC1 gene expression and reduced extracellular immune recognition. Upregulation of SQLE and CYP51 genes stimulated steroid hormone synthesis within cells, which, upon binding to G protein-coupled receptors, influenced calcium ion transport, inhibited immune activation reactions, and further reduced intracellular synthesis of anti-inflammatory substances like arachidonic acid. Ultimately, this cascade led to inflammation progression, weakened intestinal peristalsis, and villi adhesion. This study utilized multi-level omics detection to investigate the pathological symptoms of enteritis and proposed a plausible pathogenic mechanism, providing innovative insights into enteritis verification and treatment in offshore cage culture of golden pompano.
Collapse
Affiliation(s)
- Shipeng Yuan
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao Marine Science and Technology Center, Qingdao, 266003, China
| | - Minmin Sun
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Di Ma
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Xiaodan Guo
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Zhuoyu Wang
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Jingjing Niu
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Weiming Jiang
- Guangxi Academy of Fishery Sciences, No.8,Qingshan Load, Nanning, Guangxi, 530021, China
| | - Yan He
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Mingli Wei
- Guangxi Academy of Fishery Sciences, No.8,Qingshan Load, Nanning, Guangxi, 530021, China.
| | - Jie Qi
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institute / MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China.
| |
Collapse
|
50
|
Mu K, Kitts DD. Gallic acid mitigates intestinal inflammation and loss of tight junction protein expression using a 2D-Caco-2 and RAW 264.7 co-culture model. Arch Biochem Biophys 2024; 756:109978. [PMID: 38636693 DOI: 10.1016/j.abb.2024.109978] [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: 02/23/2024] [Accepted: 03/24/2024] [Indexed: 04/20/2024]
Abstract
A 2D-intestinal epithelial Caco-2/RAW 264.7 macrophage co-culture model was developed to demonstrate the relative efficacy of different phenolic acids to mitigate changes in Caco-2 epithelial cell redox state initiated both directly by autoxidation products, H2O2, and indirectly through cell communication events originating from cytokine stimulated macrophage. An inducer cocktail (lipopolysaccharide + interferon gamma) was used to activate RAW 264.7 cells in the 2D- Caco-2/RAW co-culture and intracellular changes in Caco-2 cell redox signaling occurred in response to positive changes (p < 0.05) in inflammatory biomarkers derived in macrophage that included IL-6, TNF-α, nitric oxide and peroxynitrite, respectively. Phenolic acids varied in relative capacity to reduce NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) in cocktail inflamed induced macrophage. This response in addition to the relative predisposition of gallic acid (GA) to undergo autoxidation to generate H2O2 activity (p < 0.05), culminated in downstream cell signaling in Caco-2 nuclear factor erythroid 2-related factor (Nrf2) activity (increase 26.9 %), altered monolayer integrity (increase 33.7 %), and release of interleukin 8 (IL-8) (decrease 80.5 %) (p < 0.05). It can be concluded that the co-culture model described herein was useful to assess the importance of communication between cytokine stimulated macrophage and intestinal cells. Moreover, the relative unique efficacy of GA, compared to other phenolic acids tested to protect against activated macrophage induced changes related to intestinal dysfunction were particularly relevant to epithelial redox signaling, intestinal permeability and regulation of tight junction proteins. This study concludes that phenolic acids are not equal in the capacity to protect against intestinal cell dysfunction despite some indication of biological activity.
Collapse
Affiliation(s)
- Kaiwen Mu
- Food Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, 2205 East Mall, Vancouver, V6T-1Z4, B.C, Canada
| | - David D Kitts
- Food Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, 2205 East Mall, Vancouver, V6T-1Z4, B.C, Canada.
| |
Collapse
|