1
|
Schimmele T, Langgartner D, Gebauer D, Mazzari G, Probst J, Weingast G, Ignatius A, Tabacco G, Naciu AM, Messina MV, Palermo A, Reber SO, Haffner-Luntzer M. Early life adversity promotes a milieu in favor of catabolic bone turnover in females: Mycobacterium vaccae NCTC 11659 proofs protective in preclinical studies. Brain Behav Immun 2025:S0889-1591(25)00187-4. [PMID: 40383402 DOI: 10.1016/j.bbi.2025.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
Despite early clinical studies supporting the hypothesis that early life adversity (ELA) negatively affects the bone and despite typical ELA-associated disorders, including post-traumatic stress disorder (PTSD) and major depressive disorder (MD), are associated with osteoporosis and increased bone fracture risk, preclinical studies do not support this association. However, previous studies were only performed using male and not female mice. In the current study we showed that ELA, induced by the classical maternal separation (MS) paradigm, facilitated femoral osteoclast activity specifically in female but not male mice. This was associated with a transient decline in both intestinal alpha diversity and Firmicutes/Bacteroidetes ratio, suggesting that the microbiome-gut-bone axis is involved in these effects. Moreover, ELA long-lastingly increased the femoral mRNA expression of the proinflammatory cytokine Interleukin-6 (IL-6) and the osteoclastic markers Cathepsin K and RANKL. Importantly, all sex-specific ELA effects on bone were absent in female mice administered with M. vaccae NCTC 11659 following ELA exposure. Finally, our clinical data indicate strong associations between ELA and development of an osteopenic/osteoporotic bone phenotype in postmenopausal women undergoing bone diagnostics. Together, our preclinical and clinical findings indicate that i) ELA negatively affects the bone, ii) these effects are specific for female sex, iii) the negative effects of ELA on female bone are associated with transient changes in the composition of the intestinal microbiome followed by long-lasting activation of the immune system and the HPA axis, together setting the stage for a facilitated catabolic bone turnover and development of an osteopenic/osteoporotic bone phenotype, iv) developing immunoregulatory approaches, such as repeated s.c. administrations with immunoregulatory microorganisms, have potential for prevention/treatment of ELA-related bone disorders.
Collapse
Affiliation(s)
- Tamara Schimmele
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Dorothea Gebauer
- Institute of Orthopaedic Research and Biomechanics, Ulm University Medical Center, Germany
| | - Giulia Mazzari
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Julian Probst
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Giulia Weingast
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Ulm University Medical Center, Germany
| | - Gaia Tabacco
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Anda Mihaela Naciu
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Maria Vittoria Messina
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Andrea Palermo
- Unit of Metabolic Bone and Thyroid Diseases, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany; Institute of Orthopaedic Research and Biomechanics, Ulm University Medical Center, Germany; German Center for Mental Health (DZPG), partner site Mannheim//-Heidelberg//-Ulm, Germany.
| | | |
Collapse
|
2
|
Damiani F, Giuliano MG, Cornuti S, Putignano E, Tognozzi A, Suckow V, Kalscheuer VM, Pizzorusso T, Tognini P. Multi-site investigation of gut microbiota in CDKL5 deficiency disorder mouse models: Targeting dysbiosis to improve neurological outcomes. Cell Rep 2025; 44:115546. [PMID: 40220293 PMCID: PMC12014524 DOI: 10.1016/j.celrep.2025.115546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a rare neurodevelopmental disorder often associated with gastrointestinal (GI) issues and subclinical immune dysregulation, suggesting a link to the gut microbiota. We analyze the fecal microbiota composition in two CDKL5 knockout (KO) mouse models at postnatal days (P) 25, 32 (youth), and 70 (adulthood), revealing significant microbial imbalances, particularly during juvenile stages. To investigate the role of the intestinal microbiota in CDD and assess causality, we administer antibiotics, which lead to improved visual cortical responses and reduce hyperactivity. Additionally, microglia morphology changes, indicative of altered surveillance and activation states, are reversed. Strikingly, fecal transplantation from CDKL5 KO to wild-type (WT) recipient mice successfully transfers both visual response deficits and hyperactive behavior. These findings show that gut microbiota alterations contribute to the severity of neurological symptoms in CDD, shedding light on the interplay between microbiota, microglia, and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Francesca Damiani
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Maria Grazia Giuliano
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Sara Cornuti
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Elena Putignano
- Institute of Neuroscience, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Andrea Tognozzi
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; PhD Program in Clinical and Translational Science, University of Pisa, Via Savi 10, 56126 Pisa, Italy
| | - Vanessa Suckow
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195 Berlin, Germany
| | - Vera M Kalscheuer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195 Berlin, Germany
| | - Tommaso Pizzorusso
- Laboratory of Biology BIO@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Institute of Neuroscience, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Paola Tognini
- Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Piazza Martiri della Libertà 33, 56127 Pisa, Italy.
| |
Collapse
|
3
|
Ito K, Islam J, Sakurai K, Koyama S, Matsuo A, Okano K, Hirakawa R, Furukawa M, Nochi T. Breast milk stabilizes bacterial communities in the large intestine even after weaning. Biochem Biophys Res Commun 2025; 756:151585. [PMID: 40068432 DOI: 10.1016/j.bbrc.2025.151585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
The development and maintenance of a balanced microbiota is crucial for human health. Milk contains immune factors that not only protect offspring from infectious diseases but also play an important role in promoting the development and maintenance of the microbiota. However, the persisting effects of milk-derived immune factors on the maintenance of the microbiota after weaning have not been carefully examined. In this study, a cross-fostering model was employed using immunocompetent (IC) and immunodeficient (ID) mice in which one-half of the pups born from two dams were replaced. As a result, breast milk from the IC dam (IC milk) affected the development of the microbiota during lactation and maintained it even after weaning in the large intestine of the ID pups. The large intestinal microbiota of ID pups raised on IC milk remained similar to that of normal IC pups. Under normal conditions, the genus Mucispirillum was closely associated with other bacteria, forming a diverse bacterial community in the large intestine. In the small intestine, there were no differences in the microbiota before weaning, regardless of whether IC or ID milk was consumed. By contrast, significant differences were observed in the small intestinal microbiota between IC and ID mice after weaning; however, this was dependent on the immune-related characteristics of offspring (rather than milk-derived immune factors). These results indicate that breast milk plays an important role in the large (not small) intestine of offspring to create and maintain a diverse microbiota with a balanced bacterial network even after weaning.
Collapse
Affiliation(s)
- Kaori Ito
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Jahidul Islam
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Kota Sakurai
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Saeka Koyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Graduate Program in Food Science, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | | | - Kunihiro Okano
- GENODAS Co., Ltd. Miyagi, 980-8572, Japan; Department of Biological Environment, Faculty of Bioresource Sciences, Akita Prefectural University, Akita, 010-0195, Japan
| | - Ryota Hirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Graduate Program in Food Science, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Animal Bioscience, University of Guelph, Ontario, N1G 2W1, Canada; Center for Professional Development, Institute for Excellence in Higher Education, Tohoku University, Miyagi, 980-8576, Japan.
| |
Collapse
|
4
|
Chu S, Fan R, Dai L, Liu M. Exploring the effect of soybean fermentation broth (S-FB) on gut microbes of lipopolysaccharide (LPS)-infected loach ( Misgurnus anguillicaudatus) using 16S rRNA sequencing. Front Microbiol 2025; 16:1551409. [PMID: 40170929 PMCID: PMC11958950 DOI: 10.3389/fmicb.2025.1551409] [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: 12/25/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
The fermentation products of soybean are rich in beneficial bacteria, which play Shenghui Chu a significant role in maintaining the balance of intestinal microbiota and improving intestinal health. To investigate the immunomodulatory effects of soybean fermentation broth (S-FB) on loach (Misgurnus anguillicaudatus) with lipopolysaccharide (LPS)-induced enteritis, 16S rDNA high-throughput sequencing technology was employed to analyze the composition and structure of intestinal microbiota in two groups: the LPS-treated group (fed with soybean broth) and the control group (normal feeding conditions). The results revealed that the relative abundance of beneficial bacteria, such as Lactobacillus and Muribaculaceae, significantly increased in the treatment group, while the relative abundance of harmful bacteria, including Aeromonas and Shewanella, decreased. These findings suggest that soybean fermentation broth can repair intestinal damage and maintain intestinal health by enhancing the abundance of beneficial bacteria and reducing the pathogenic effects of harmful bacteria on the host. Functional prediction studies of microbial communities also showed that treatment groups primarily affected metabolic and genetic information processing. The research results analyzed the changes in the structure and distribution of intestinal microflora in different groups of loach, providing new insights into the possible role of soybean fermentation liquid in intestinal inflammation.
Collapse
Affiliation(s)
- Shenghui Chu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, School of Pharmacy, Ministry of Education, Shihezi University, Shihezi, China
| | - Ruike Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, China
| | - Lishang Dai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- School of Traditional Chinese Medicine, Wenzhou Medical University, Wenzhou, China
| | - Min Liu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, School of Pharmacy, Ministry of Education, Shihezi University, Shihezi, China
| |
Collapse
|
5
|
Mahdavi M, Kim TY, Prévost K, Balthazar P, Gagné-Ouellet V, Hus IFP, Duchesne É, Harvey S, Gagnon C, Laforest-Lapointe I, Dumont NA, Massé E. Influence of CTG repeats from the human DM1 locus on murine gut microbiota. Comput Struct Biotechnol J 2025; 27:733-743. [PMID: 40092662 PMCID: PMC11908463 DOI: 10.1016/j.csbj.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Myotonic Dystrophy type 1 (DM1) is caused by a CTG repeat expansion in the 3' untranslated region of the DMPK gene. This expansion leads to the production of toxic RNA transcripts, which accumulate in the nucleus and interfere with normal RNA processing. DM1 affects a broad range of tissues and systems such as the skeletal muscle, the central nervous system, cardiac, visual, reproductive, and gastrointestinal (GI) system. GI dysfunction is a significant but poorly understood aspect of DM1. Particularly, it is unknown if there are alterations in the intestinal microbiome in DM1. Here, we used a transgenic humanized mouse model (DMSXL) to explore how the gut microbiome may be linked to GI issues in DM1. For this purpose, 68 stool samples from Homozygous, Heterozygous, and Wild-Type (WT) mice were collected. These samples were sequenced by MiSeq and analyzed with DADA2 to generate taxonomic profiles. Our analysis indicated that the overexpression of CTG repeats significantly influences the bacterial structure of the gut microbiome in Homozygous mice samples, especially in terms of the relative abundance of the Patescibacteria and Defferibacterota Phyla. These results provide valuable information about the gut microbiota structure thus improving the understanding of the role of these changes in the pathogenicity as well as GI problems of DM1 patients.
Collapse
Affiliation(s)
- Manijeh Mahdavi
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| | - Tae-Yeon Kim
- Department of microbiology, infectiology and immunology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Karine Prévost
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| | - Philippe Balthazar
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| | - Valérie Gagné-Ouellet
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Isabelle Fissette-Paul Hus
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Élise Duchesne
- School of Rehabilitation Sciences, Faculty of Medicine, Université Laval, Quebec, Quebec, Canada
- Neuromuscular Diseases Interdisciplinary Research Group (GRIMN), Saguenay-Lac-St-Jean Integrated University Health and Social Services Center, Saguenay, Quebec, Canada
- Center for Interdisciplinary Research in Rehabilitation and Social Integration (Cirris), Capitale-Nationale Integrated University Health and Social Services Center, Quebec, Quebec, Canada
- CHU de Québec - Université Laval Research Center, Québec, Québec, Canada
| | - Séréna Harvey
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Cynthia Gagnon
- Department of Rehabilitation, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Isabelle Laforest-Lapointe
- Départment de Biologie, Faculté des Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
- Centre de Recherche du Centre Hospitalier universitaire de Sherbrooke (CR-CHUS), Sherbrooke, QC J1H 5N4, Canada
| | - Nicolas A. Dumont
- CHU Sainte-Justine Research Center, Montréal, QC, Canada
- School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Eric Massé
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, QC J1E 4K8, Canada
| |
Collapse
|
6
|
Chen F, Liu Z, Xie C, He J, Chen J, Peng K, Chen X, He J, Liu Z, Yang H, Kang K, He B, Lin Q. The effect of Alpinia oxyphylla essential oil on growth performance, immune, antioxidant functions and gut microbiota in pigs. Front Vet Sci 2024; 11:1468520. [PMID: 39720412 PMCID: PMC11666522 DOI: 10.3389/fvets.2024.1468520] [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: 07/22/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024] Open
Abstract
Alpinia oxyphylla, a perennial herb belonging to the Zingiberaceae family, has a long history of traditional medicinal use. The present study evaluated the efficacy of different concentrations of Alpinia oxyphylla essential oil (AEO) on the growth performance, serum antioxidation capacities, immune function, apparent digestibility of nutrients, and gut microbiota in fattening pigs. A total of 120 pigs were divided into five treatments, with six replicates each and four pigs per replicate. The pigs were fed a basal diet or basal diet with chlortetracycline (CTC) alone or AEO at 250, 500, and 1,000 mg/kg (referred to as groups AEO1, AEO2, and AEO3, respectively) for 35 days, preceded by a 7-day pre-feed period. The results show that there were no statistically significant differences in growth performance for any dose of AEO supplementation. AEO increased L-DLC content, total protein content and the activity of GSH in serum (p < 0.05). The AEO also exhibited a linear increase in serum IgG content (p < 0.05). Dietary supplementation with AEO improved apparent digestibility of crude ash and calcium (p < 0.05). In gut microbiota, AEO modified the diversity and abundance of bacterial communities in fattening pigs. The abundance of Dorea, Blautia, Butyricicoccus, Bulleidia, and Lactobacillus was higher in the AEO groups compared to the control group, while Clostridium and Turicibacter were lower. The Bifidobacteriales and Pseudomonas were abundant in group AEO1 and AEO3, respectively. In conclusion, dietary supplementation of 1,000 mg/kg AEO has the potential to improve growth performance, immunological, biochemical, and antioxidant statuses. Additionally, AEO can increase the efficiency of nutrient digestion and absorption through the regulation of gut microbiota.
Collapse
Affiliation(s)
- Fengming Chen
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Zhimou Liu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
- Hunan Nuoz Biological Technology Co., Ltd., Yiyang, Hunan, China
| | - Chun Xie
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jieyi He
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Jiayi Chen
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Kaiqiang Peng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xu Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jiajia He
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Zhenyi Liu
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Hui Yang
- Hunan Academy of Agricultural Sciences, Changsha, China
| | - Kelang Kang
- Hunan Academy of Agricultural Sciences, Changsha, China
| | - Binsheng He
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
| | - Qian Lin
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, Hunan, China
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| |
Collapse
|
7
|
Liang Y, Zhang C, Xiong X, Mao X, Sun P, Yue Z, Wang W, Xie R, Zhang G. Alterations of gut microbiome in eosinophilic chronic rhinosinusitis. Eur Arch Otorhinolaryngol 2024; 281:6459-6468. [PMID: 39212702 PMCID: PMC11564334 DOI: 10.1007/s00405-024-08931-3] [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: 03/30/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE A growing body of evidence has elucidated that the gut microbiota has a crucial impact on the pathophysiological process of atopic diseases. Eosinophilic chronic rhinosinusitis with nasal polyps (eCRSwNP) is a local atopic disease of the systemic immune response. Alterations in the gut microbiome in eCRSwNP patients remain largely undefined. METHODS 16S rRNA gene sequencing was performed in a cross-sectional study of 17 eCRSwNP patients, 9 noneCRSwNP patients and 13 healthy controls, and gut microbiota DNA sequencing between each pair of groups was compared using bioinformatic methods. RESULTS Compared with that of healthy controls, the gut microbiomes of eCRSwNP patients were characterised by a distinct overall microbial composition. However, no significant differences were found in the alpha diversity of the gut microbiota between patients and healthy controls. The distinct differences in microbial composition were significantly correlated with the severity of disease. At the genus level, the abundance of Faecalibacterium positively correlated with Lund-Mackay CT scores. Similarly, the abundance of Turicibacter positively correlated with the percentage of tissue eosinophils. CONCLUSIONS We found alterations in the gut microbiome in eCRSwNP patients, and the alterations in the gut microbiome were correlated with the severity of disease.
Collapse
Affiliation(s)
- Yibo Liang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Chenting Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Xin Xiong
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Xiang Mao
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Peiyong Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Zhenzhong Yue
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China
| | - Runxiang Xie
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, 59 Liu Ting Street, Haishu District, Ningbo, 315000, Zhejiang, China.
| | - Guimin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Institute of Otolaryngology of Tianjin, Key Laboratory of Auditory Speech and Balance Medicine, Key Medical Discipline of Tianjin (Otolaryngology), Tianjin First Central Hospital, Quality Control Centre of Otolaryngology, Tianjin, 300192, China.
| |
Collapse
|
8
|
Li X, Wang Q, Wang F, Jin Q, Deng B, Yang R, Fu A, Li F, Zhang Q, Li W. Rosa roxburghii Tratt (Cili) has a more effective capacity in alleviating DSS-induced colitis compared to Vitamin C through B cell receptor pathway. Food Res Int 2024; 195:114950. [PMID: 39277228 DOI: 10.1016/j.foodres.2024.114950] [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: 04/12/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024]
Abstract
Rosa roxburghii Tratt (RRT), a traditional Chinese plant known as the 'King of Vitamin C (VitC; ascorbic acid, AsA)', contains a wealth of nutrients and functional components, including polysaccharides, organic acids, flavonoids, triterpenes, and high superoxide dismutase (SOD) activity. The various functional components of RRT suggest that it may theoretically have a stronger potential for alleviating colitis compared to VitC. This study aims to verify whether RRT has a stronger ability to alleviate colitis than equimolar doses of VitC and to explore the mechanisms underlying this improvement. Results showed that RRT significantly mitigated body weight loss, intestinal damage, elevated inflammation levels, and compromised barriers in mice induced by Dextran sulfate sodium (DSS). Additionally, RRT enhanced the diversity and composition of intestinal microbiota in these DSS-induced mice. Colon RNA sequencing analysis revealed that compared to VitC, RRT further downregulated multiple immune-related signaling pathways, particularly the B cell receptor (BCR) pathway, which is centered around genes like Btk and its downstream PI3K-AKT, NF-κB, and MAPK signaling pathways. Correlation analysis between microbiota and genes demonstrated a significant relationship between the taxa improved by RRT and the key genes in the BCR and its downstream signaling pathways. Overall, RRT exhibited superior capabilities in alleviating DSS-induced colitis compared to VitC by decreasing intestinal inflammation and modulating BCR and its downstream signaling pathways, potentially regulated by the improved intestinal microbiota.
Collapse
Affiliation(s)
- Xiang Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Qi Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Fei Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Qian Jin
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Bin Deng
- Guizhou Light Industry Technical College, Guiyang 550025, Guizhou Province, China
| | - RongChang Yang
- Nanjing Kangyou Biotechnology Co., Ltd., Nanjing 211316, Jiangsu Province, China
| | - Aikun Fu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Fuyong Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Qiao Zhang
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Research Center for Clinical Pharmacy, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China.
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Animal Nutrition and Feed Sciences, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China; Zhejiang Youheyhey Biotechnology Co., LTD, Huzhou 313000, Zhejiang Province, China.
| |
Collapse
|
9
|
Kang M, Kang M, Lee J, Yoo J, Lee S, Oh S. Allium tuberosum-derived nanovesicles with anti-inflammatory properties prevent DSS-induced colitis and modify the gut microbiome. Food Funct 2024; 15:7641-7657. [PMID: 38953279 DOI: 10.1039/d4fo01366b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Edible plant-derived nanovesicles (ePDNs) have shown potential as a non-pharmacological option for inflammatory bowel disease (IBD) by maintaining gut health and showing anti-inflammatory effects. However, the effects of Allium tuberosum-derived nanovesicles (ADNs) on colitis have not been studied to date. Here, we extracted exosome-like nanovesicles from Allium tuberosum and investigated whether they have an anti-inflammatory effect in RAW 264.7 cells and colitis mice. The results showed that ADNs reduced the elevated levels of inflammatory factors such as IL-1β, IL-6, TNF-α, and NF-κB pathway-related proteins as a consequence of lipopolysaccharide (LPS) stimulation in RAW 264.7 cells. Furthermore, our mouse experiments demonstrated that ADNs could ameliorate dextran sulfate sodium (DSS)-induced colitis symptoms (e.g., increased disease activity index score, intestinal permeability, and histological appearance). Additionally, ADNs counteracted DSS-induced colitis by downregulating the expression of serum amyloid A (SAA), IL-1β, IL-6, and TNF-α and increasing the expression of tight junction proteins (ZO-1 and occludin) and the anti-inflammatory cytokine IL-10. 16S rRNA gene sequencing showed that ADN intervention restored the gut microbial composition, which was similar to that of the DSS non-treated group, by decreasing the ratio of Firmicutes to Bacteroidetes and the relative abundance of Proteobacteria. Furthermore, ADNs induced acetic acid production along with an increase in the abundance of Lactobacillus. Overall, our findings suggest that ADN supplementation has a crucial role in maintaining gut health and is a novel preventive therapy for IBD.
Collapse
Affiliation(s)
- Minkyoung Kang
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Minji Kang
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Juyeon Lee
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Jiseon Yoo
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Sujeong Lee
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
| | - Sangnam Oh
- Department of Environmental Science and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea.
- Department of Food and Nutrition, Jeonju University, Jeonju 55069, Republic of Korea
| |
Collapse
|
10
|
Evans L, Barral P. CD1 molecules: Beyond antigen presentation. Mol Immunol 2024; 170:1-8. [PMID: 38579449 PMCID: PMC11481681 DOI: 10.1016/j.molimm.2024.03.011] [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: 09/28/2022] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
CD1 molecules are well known for their role in binding and presenting lipid antigens to mediate the activation of CD1-restricted T cells. However, much less appreciated is the fact that CD1 molecules can have additional "unconventional" roles which impact the activation and functions of CD1-expressing cells, ultimately controlling tissue homeostasis as well as the progression of inflammatory and infectious diseases. Some of these roles are mediated by so-called reverse signalling, by which crosslinking of CD1 molecules at the cell surface initiates intracellular signalling. On the other hand, CD1 molecules can also control metabolic and inflammatory pathways in CD1-expressing cells through cell-intrinsic mechanisms independent of CD1 ligation. Here, we review the evidence for "unconventional" functions of CD1 molecules and the outcomes of such roles for health and disease.
Collapse
Affiliation(s)
- Lauren Evans
- The Peter Gorer Department of Immunobiology. King's College London, London, UK; The Francis Crick Institute, London, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology. King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
11
|
Maritan E, Quagliariello A, Frago E, Patarnello T, Martino ME. The role of animal hosts in shaping gut microbiome variation. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230071. [PMID: 38497257 PMCID: PMC10945410 DOI: 10.1098/rstb.2023.0071] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 03/19/2024] Open
Abstract
Millions of years of co-evolution between animals and their associated microbial communities have shaped and diversified the nature of their relationship. Studies continue to reveal new layers of complexity in host-microbe interactions, the fate of which depends on a variety of different factors, ranging from neutral processes and environmental factors to local dynamics. Research is increasingly integrating ecosystem-based approaches, metagenomics and mathematical modelling to disentangle the individual contribution of ecological factors to microbiome evolution. Within this framework, host factors are known to be among the dominant drivers of microbiome composition in different animal species. However, the extent to which they shape microbiome assembly and evolution remains unclear. In this review, we summarize our understanding of how host factors drive microbial communities and how these dynamics are conserved and vary across taxa. We conclude by outlining key avenues for research and highlight the need for implementation of and key modifications to existing theory to fully capture the dynamics of host-associated microbiomes. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Elisa Maritan
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| | - Andrea Quagliariello
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| | - Enric Frago
- CIRAD, UMR CBGP, INRAE, Institut Agro, IRD, Université Montpellier, 34398 Montpellier, France
| | - Tomaso Patarnello
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| | - Maria Elena Martino
- Department of Comparative Biomedicine and Food Science, University of Padova, 35020 Padova, Italy
| |
Collapse
|
12
|
Szántó M, Yélamos J, Bai P. Specific and shared biological functions of PARP2 - is PARP2 really a lil' brother of PARP1? Expert Rev Mol Med 2024; 26:e13. [PMID: 38698556 PMCID: PMC11140550 DOI: 10.1017/erm.2024.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024]
Abstract
PARP2, that belongs to the family of ADP-ribosyl transferase enzymes (ART), is a discovery of the millennium, as it was identified in 1999. Although PARP2 was described initially as a DNA repair factor, it is now evident that PARP2 partakes in the regulation or execution of multiple biological processes as inflammation, carcinogenesis and cancer progression, metabolism or oxidative stress-related diseases. Hereby, we review the involvement of PARP2 in these processes with the aim of understanding which processes are specific for PARP2, but not for other members of the ART family. A better understanding of the specific functions of PARP2 in all of these biological processes is crucial for the development of new PARP-centred selective therapies.
Collapse
Affiliation(s)
- Magdolna Szántó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - José Yélamos
- Hospital del Mar Research Institute, Barcelona, Spain
| | - Péter Bai
- HUN-REN-UD Cell Biology and Signaling Research Group, Debrecen, 4032, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary
| |
Collapse
|
13
|
Lin Q, Kuypers M, Baglaenko Y, Cao E, Hezaveh K, Despot T, de Amat Herbozo C, Cruz Tleugabulova M, Umaña JM, McGaha TL, Philpott DJ, Mallevaey T. The intestinal microbiota modulates the transcriptional landscape of iNKT cells at steady-state and following antigen exposure. Mucosal Immunol 2024; 17:226-237. [PMID: 38331095 DOI: 10.1016/j.mucimm.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Invariant Natural Killer T (iNKT) cells are unconventional T cells that respond to microbe-derived glycolipid antigens. iNKT cells exert fast innate effector functions that regulate immune responses in a variety of contexts, including during infection, cancer, or inflammation. The roles these unconventional T cells play in intestinal inflammation remain poorly defined and vary based on the disease model and species. Our previous work suggested that the gut microbiota influenced iNKT cell functions during dextran sulfate sodium-induced colitis in mice. This study, shows that iNKT cell homeostasis and response following activation are altered in germ-free mice. Using prenatal fecal transplant in specific pathogen-free mice, we show that the transcriptional signatures of iNKT cells at steady state and following αGC-mediated activation in vivo are modulated by the microbiota. Our data suggest that iNKT cells sense the microbiota at homeostasis independently of their T cell receptors. Finally, iNKT cell transcriptional signatures are different in male and female mice. Collectively, our findings suggest that sex and the intestinal microbiota are important factors that regulate iNKT cell homeostasis and responses. A deeper understanding of microbiota-iNKT cell interactions and the impact of sex could improve the development of iNKT cell-based immunotherapies.
Collapse
Affiliation(s)
- Qiaochu Lin
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Meggie Kuypers
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yuriy Baglaenko
- Center for Autoimmune Genomics and Etiology, Division of Genetics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Eric Cao
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Kebria Hezaveh
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Tijana Despot
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Tracy L McGaha
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Yu J, Chen Y, Wang J, Wu H. Research progress on the relationship between traumatic brain injury and brain-gut-microbial axis. IBRAIN 2024; 10:477-487. [PMID: 39691426 PMCID: PMC11649388 DOI: 10.1002/ibra.12153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 12/19/2024]
Abstract
Traumatic brain injury (TBI) is a common disease with a high rate of death and disability, which poses a serious threat to human health; thus, the effective treatment of TBI has been a high priority. The brain-gut-microbial (BGM) axis, as a bidirectional communication network for information exchange between the brain and gut, plays a crucial role in neurological diseases. This article comprehensively explores the interrelationship between the BGM axis and TBI, including its physiological effects, basic pathophysiology, and potential therapeutic strategies. It highlights how the bidirectional regulatory pathways of the BGM axis could provide new insights into clinical TBI treatment and underscores the necessity for advanced research and development of innovative clinical treatments for TBI.
Collapse
Affiliation(s)
- Jie Yu
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yun‐Xin Chen
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Jin‐Wei Wang
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Hai‐Tao Wu
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
15
|
Satoh M, Iwabuchi K. Contribution of NKT cells and CD1d-expressing cells in obesity-associated adipose tissue inflammation. Front Immunol 2024; 15:1365843. [PMID: 38426085 PMCID: PMC10902011 DOI: 10.3389/fimmu.2024.1365843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Natural killer T (NKT) cell are members of the innate-like T lymphocytes and recognizes lipid antigens presented by CD1d-expressing cells. Obesity-associated inflammation in adipose tissue (AT) leads to metabolic dysfunction, including insulin resistance. When cellular communication is properly regulated among AT-residing immune cells and adipocytes during inflammation, a favorable balance of Th1 and Th2 immune responses is achieved. NKT cells play crucial roles in AT inflammation, influencing the development of diet-induced obesity and insulin resistance. NKT cells interact with CD1d-expressing cells in AT, such as adipocytes, macrophages, and dendritic cells, shaping pro-inflammatory or anti-inflammatory microenvironments with distinct characteristics depending on the antigen-presenting cells. Additionally, CD1d may be involved in the inflammatory process independently of NKT cells. In this mini-review, we provide a brief overview of the current understanding of the interaction between immune cells, focusing on NKT cells and CD1d signaling, which control AT inflammation both in the presence and absence of NKT cells. We aim to enhance our understanding of the mechanisms of obesity-associated diseases.
Collapse
Affiliation(s)
- Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Japan
| | | |
Collapse
|
16
|
Lee SW, Park HJ, Van Kaer L, Hong S. Role of CD1d and iNKT cells in regulating intestinal inflammation. Front Immunol 2024; 14:1343718. [PMID: 38274786 PMCID: PMC10808723 DOI: 10.3389/fimmu.2023.1343718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Invariant natural killer T (iNKT) cells, a subset of unconventional T cells that recognize glycolipid antigens in a CD1d-dependent manner, are crucial in regulating diverse immune responses such as autoimmunity. By engaging with CD1d-expressing non-immune cells (such as intestinal epithelial cells and enterochromaffin cells) and immune cells (such as type 3 innate lymphoid cells, B cells, monocytes and macrophages), iNKT cells contribute to the maintenance of immune homeostasis in the intestine. In this review, we discuss the impact of iNKT cells and CD1d in the regulation of intestinal inflammation, examining both cellular and molecular factors with the potential to influence the functions of iNKT cells in inflammatory bowel diseases such as Crohn's disease and ulcerative colitis.
Collapse
Affiliation(s)
- Sung Won Lee
- Department of Biomedical Laboratory Science, College of Health and Biomedical Services, Sangji University, Wonju, Republic of Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Parihar N, Bhatt LK. The emerging paradigm of Unconventional T cells as a novel therapeutic target for celiac disease. Int Immunopharmacol 2023; 122:110666. [PMID: 37473709 DOI: 10.1016/j.intimp.2023.110666] [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: 04/03/2023] [Revised: 07/07/2023] [Accepted: 07/16/2023] [Indexed: 07/22/2023]
Abstract
Celiac disease (CD) is an organ-specific autoimmune disorder that occurs in genetically predisposed individuals when exposed to exogenous dietary gluten. This exposure to wheat gluten and related proteins from rye and barley triggers an immune response which leads to the development of enteropathy associated with symptoms of bloating, diarrhea, or malabsorption. The sole current treatment is to follow a gluten-free diet for the rest of one's life. Intestinal barriers are enriched with Unconventional T cells such as iNKT, MAIT, and γδ T cells, which lack or express only a limited range of rearranged antigen receptors. Unconventional T cells play a crucial role in regulating mucosal barrier function and microbial colonization. Unconventional T cell populations are widely represented in diseased conditions, where changes in disease activity related to iNKT and MAIT cell reduction, as well as γδ T cell expansion, are demonstrated. In this review, we discuss the role and potential employment of Unconventional T cells as a therapeutic target in the pathophysiology of celiac disease.
Collapse
Affiliation(s)
- Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
18
|
Desjardins A, Zerfas P, Filion D, Palmer RJ, Falcone EL. Mucispirillum schaedleri: Biofilm Architecture and Age-Dependent Pleomorphy. Microorganisms 2023; 11:2200. [PMID: 37764045 PMCID: PMC10535455 DOI: 10.3390/microorganisms11092200] [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: 07/21/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Round bodies in spirochete cultures have been a controversial subject since their description seven decades ago. We report the existence of round bodies (spherical cells) in cultures of Mucispirillum schaedleri, a spiral bacterium phylogenetically distant from spirochetes. Furthermore, when grown in biofilms, M. schaedleri demonstrates a unique morphology known as cording, which has been previously described only in mycobacteria. Thus, M. schaedleri has two distinct features, each previously thought to be unique to two different phylogenetically distant groups of bacteria.
Collapse
Affiliation(s)
- Aléhandra Desjardins
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC H2W 1R7, Canada;
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Patricia Zerfas
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Dominic Filion
- Microscopy and Imaging Platform, Montreal Clinical Research Institute (IRCM), Montreal, QC H2W 1R7, Canada
| | - Robert J. Palmer
- National Institute of Dental and Craniofacial Research, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Emilia Liana Falcone
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute (IRCM), Montreal, QC H2W 1R7, Canada;
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
19
|
Luo J, Chen Z, Castellano D, Bao B, Han W, Li J, Kim G, An D, Lu W, Wu C. Lipids regulate peripheral serotonin release via gut CD1d. Immunity 2023; 56:1533-1547.e7. [PMID: 37354904 PMCID: PMC10527042 DOI: 10.1016/j.immuni.2023.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/04/2023] [Accepted: 06/01/2023] [Indexed: 06/26/2023]
Abstract
The crosstalk between the immune and neuroendocrine systems is critical for intestinal homeostasis and gut-brain communications. However, it remains unclear how immune cells participate in gut sensation of hormones and neurotransmitters release in response to environmental cues, such as self-lipids and microbial lipids. We show here that lipid-mediated engagement of invariant natural killer T (iNKT) cells with enterochromaffin (EC) cells, a subset of intestinal epithelial cells, promoted peripheral serotonin (5-HT) release via a CD1d-dependent manner, regulating gut motility and hemostasis. We also demonstrated that inhibitory sphingolipids from symbiotic microbe Bacteroides fragilis represses 5-HT release. Mechanistically, CD1d ligation on EC cells transduced a signal and restrained potassium conductance through activation of protein tyrosine kinase Pyk2, leading to calcium influx and 5-HT secretion. Together, our data reveal that by engaging with iNKT cells, gut chemosensory cells selectively perceive lipid antigens via CD1d to control 5-HT release, modulating intestinal and systemic homeostasis.
Collapse
Affiliation(s)
- Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - David Castellano
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Bin Bao
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Girak Kim
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Dingding An
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
20
|
Tu R, Zhou C, Huang W, Feng Z, Zhao Q, Shi X, Cui L, Chen K. Fuzi polysaccharides improve immunity in immunosuppressed mouse models by regulating gut microbiota composition. Heliyon 2023; 9:e18244. [PMID: 37519691 PMCID: PMC10372400 DOI: 10.1016/j.heliyon.2023.e18244] [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: 10/18/2022] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023] Open
Abstract
Rationale and objectives Fuzi, the dried root of Aconitum carmichaelii Debx, is one of the widely used traditional Chinese medicines. Fuzi polysaccharides are considered the most bioactive compounds with immunomodulatory functions, however, the mechanisms have not been evaluated. This study aims to systematically investigate the effects of Fuzi polysaccharides on the gut microbiota and immune function using a mouse model immunosuppressed with cyclophosphamide. Methods The short-chain fatty acid levels in cecal contents were measured by gas chromatography-mass spectrometry. The gut microbiota 16S rRNA gene were sequenced by next generation sequencing. The mRNA expression levels of NF-κB, IL-6, TNF-α, iNOS and COX-2 were measured using quantitative real-time polymerase chain reaction. The protein expression of occludin and zonula occludens-1 were analyzed by Western blot. The white blood cells were counted using automated hematology analyzer, and CD4+FOXP3+/CD4+ ratio was measured by flow cytometry. Results and Conclusions Fuzi polysaccharides had the function of elevating the concentration of acetic acid, propionic acid, isobutyric acid, and n-butyric acid in the cecum. Meanwhile, Fuzi polysaccharides could decrease the relative abundance of Helicobacter, Anaerotruncus, Faecalibacterium, Lachnospira, Erysipelotrichaceae_UCG-003, Mucispirillum, and Mycoplasma, and increase the relative abundance of Rhodospirillales, Ruminococcaceae_UCG-013, Mollicutes_RF39, Ruminococcus_1, Christensenellaceae_R-7_group, and Muribaculaceae in the gut. Furthermore, Fuzi polysaccharides exhibited the function of increasing spleen and thymus indices and number of white blood cells and lymphocytes. Fuzi polysaccharides could reverse the decreased mRNA expression of NF-кB, IL-6, and iNOS, differentiation of CD4+FOXP3+ regulatory T cells as well as protein expression of occludin and zonula occludens-1 induced by cyclophosphamide. In addition, the mRNA and protein expression of cytokines were significantly correlated with the abundance of gut microbiota under Fuzi polysaccharides treatment. Collectively, the above results demonstrated that Fuzi polysaccharides could regulate inflammatory cytokines and gut microbiota composition of immunosuppressive mice to improve immunity, thereby shedding light on revealing the molecular mechanism of polysaccharides of traditional Chinese medicines in the future.
Collapse
Affiliation(s)
- Ran Tu
- Medical Laboratory of Jingmen People's Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | - Cheng Zhou
- Medical Laboratory of Jingmen People's Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | - Wenfeng Huang
- Medical Laboratory of Jingmen People's Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | - Zhengping Feng
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
- Yan'an Hospital of Traditional Chinese Medicine, Yan'an, Shaanxi, China
| | - Qiufang Zhao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xiaofei Shi
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Langjun Cui
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Keke Chen
- School of Biological and Environmental Engineering, Xi'an University, Xi'an Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an, Shaanxi, China
| |
Collapse
|
21
|
Van Pee T, Nawrot TS, van Leeuwen R, Hogervorst J. Ambient particulate air pollution and the intestinal microbiome; a systematic review of epidemiological, in vivo and, in vitro studies. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:162769. [PMID: 36907413 DOI: 10.1016/j.scitotenv.2023.162769] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 05/13/2023]
Abstract
A healthy indigenous intestinal microbiome is indispensable for intra- and extra-intestinal human health. Since well-established factors such as diet and antibiotic use only explain 16 % of the inter-individual variation in gut microbiome composition, recent studies have focused on the association between ambient particulate air pollution and the intestinal microbiome. We systematically summarize and discuss all evidence concerning the effect of particulate air pollution on intestinal bacterial diversity indices, specific bacterial taxa, and potential underlying intestinal mechanisms. To this end, all possibly relevant publications published between February 1982 and January 2023 were screened, and eventually, 48 articles were included. The vast majority (n = 35) of these studies were animal studies. The exposure periods investigated in the human epidemiological studies (n = 12) ranged from infancy through elderly. This systematic review found that intestinal microbiome diversity indices were generally negatively associated with particulate air pollution in epidemiological studies, with an increase in taxa belonging to Bacteroidetes (two studies), Deferribacterota (one study), and Proteobacteria (four studies), a decrease in taxa belonging to Verrucomicrobiota (one study), and no consensus for taxa belonging to Actinobacteria (six studies) and Firmicutes (seven studies). There was no unequivocal effect of ambient particulate air pollution exposure on bacterial indices and taxa in animal studies. Only one study in humans examined a possible underlying mechanism; yet, the included in vitro and animal studies depicted higher gut damage, inflammation, oxidative stress, and permeability in exposed versus unexposed animals. Overall, the population-based studies showed a dose-related continuum of short- and long-term ambient particulate air pollution exposure on lower gut diversity and shifts in taxa over the entire life course.
Collapse
Affiliation(s)
- Thessa Van Pee
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590 Diepenbeek, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590 Diepenbeek, Belgium; Department of Public Health and Primary Care, Leuven University, Herestraat 49-box 706, 3000 Leuven, Belgium.
| | - Romy van Leeuwen
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590 Diepenbeek, Belgium
| | - Janneke Hogervorst
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590 Diepenbeek, Belgium
| |
Collapse
|
22
|
Joyce S, Okoye GD, Driver JP. Die Kämpfe únd schláchten-the struggles and battles of innate-like effector T lymphocytes with microbes. Front Immunol 2023; 14:1117825. [PMID: 37168859 PMCID: PMC10165076 DOI: 10.3389/fimmu.2023.1117825] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/22/2023] [Indexed: 05/13/2023] Open
Abstract
The large majority of lymphocytes belong to the adaptive immune system, which are made up of B2 B cells and the αβ T cells; these are the effectors in an adaptive immune response. A multitudinous group of lymphoid lineage cells does not fit the conventional lymphocyte paradigm; it is the unconventional lymphocytes. Unconventional lymphocytes-here called innate/innate-like lymphocytes, include those that express rearranged antigen receptor genes and those that do not. Even though the innate/innate-like lymphocytes express rearranged, adaptive antigen-specific receptors, they behave like innate immune cells, which allows them to integrate sensory signals from the innate immune system and relay that umwelt to downstream innate and adaptive effector responses. Here, we review natural killer T cells and mucosal-associated invariant T cells-two prototypic innate-like T lymphocytes, which sense their local environment and relay that umwelt to downstream innate and adaptive effector cells to actuate an appropriate host response that confers immunity to infectious agents.
Collapse
Affiliation(s)
- Sebastian Joyce
- Department of Veterans Affairs, Tennessee Valley Healthcare Service, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, The Vanderbilt Institute for Infection, Immunology and Inflammation and Vanderbilt Center for Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gosife Donald Okoye
- Department of Pathology, Microbiology and Immunology, The Vanderbilt Institute for Infection, Immunology and Inflammation and Vanderbilt Center for Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John P. Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
23
|
Acetylated xylo-oligosaccharide from Hawthorn kernels inhibits colon cancer cells in vitro and in vivo. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
24
|
Nan Q, Ye Y, Tao Y, Jiang X, Miao Y, Jia J, Miao J. Alterations in metabolome and microbiome signatures provide clues to the role of antimicrobial peptide KT2 in ulcerative colitis. Front Microbiol 2023; 14:1027658. [PMID: 36846795 PMCID: PMC9947474 DOI: 10.3389/fmicb.2023.1027658] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/10/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Ulcerative colitis (UC) is an inflammatory disease of the intestinal tract with unknown etiology. Both genetic and environmental factors are involved in the occurrence and development of UC. Understanding changes in the microbiome and metabolome of the intestinal tract is crucial for the clinical management and treatment of UC. Methods Here, we performed metabolomic and metagenomic profiling of fecal samples from healthy control mice (HC group), DSS (Dextran Sulfate Sodium Salt) -induced UC mice (DSS group), and KT2-treated UC mice (KT2 group). Results and Discussion In total, 51 metabolites were identified after UC induction, enriched in phenylalanine metabolism, while 27 metabolites were identified after KT2 treatment, enriched in histidine metabolism and bile acid biosynthesis. Fecal microbiome analysis revealed significant differences in nine bacterial species associated with the course of UC, including Bacteroides, Odoribacter, and Burkholderiales, which were correlated with aggravated UC, and Anaerotruncus, Lachnospiraceae, which were correlated with alleviated UC. We also identified a disease-associated network connecting the above bacterial species with UC-associated metabolites, including palmitoyl sphingomyelin, deoxycholic acid, biliverdin, and palmitoleic acid. In conclusion, our results indicated that Anaerotruncus, Lachnospiraceae, and Mucispirillum were protective species against DSS-induced UC in mice. The fecal microbiomes and metabolomes differed significantly among the UC mice and KT2-treated and healthy-control mice, providing potential evidence for the discovery of biomarkers of UC.
Collapse
Affiliation(s)
- Qiong Nan
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yunnan Province Clinical Research Center for Digestive Diseases, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yan Ye
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yunnan Province Clinical Research Center for Digestive Diseases, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yan Tao
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yunnan Province Clinical Research Center for Digestive Diseases, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xinyi Jiang
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yunnan Province Clinical Research Center for Digestive Diseases, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yinglei Miao
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yunnan Province Clinical Research Center for Digestive Diseases, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yinglei Miao,
| | - Jie Jia
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China,Jie Jia,
| | - Jiarong Miao
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,Yunnan Province Clinical Research Center for Digestive Diseases, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China,*Correspondence: Jiarong Miao,
| |
Collapse
|
25
|
Zhou X, Zhang X, Zhao N, Zhang L, Qiu W, Song C, Chai J, Cai S, Chen W. Gut Microbiota Deficiency Exacerbates Liver Injury in Bile Duct Ligated Mice via Inflammation and Lipid Metabolism. Int J Mol Sci 2023; 24:3180. [PMID: 36834588 PMCID: PMC9960910 DOI: 10.3390/ijms24043180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Bile components play a critical role in maintaining gut microbiota homeostasis. In cholestasis, bile secretion is impaired, leading to liver injury. However, it remains to be elucidated whether gut microbiota plays a role in cholestatic liver injury. Here, we performed a sham operation and bile duct ligation (BDL) in antibiotic-induced microbiome depleted (AIMD) mice and assessed liver injury and fecal microbiota composition in these mice. Significant reductions in gut microbiota richness and diversity were found in AIMD-sham mice when compared to sham controls. Three-day BDL leads to great elevation of plasma ALT, ALP, total bile acids, and bilirubin where reduced diversity of the gut microbiota was also found. AIMD further aggravated cholestatic liver injury evidenced by significantly higher levels of plasma ALT and ALP, associated with further reduced diversity and increased Gram-negative bacteria in gut microbiota. Further analyses revealed increased levels of LPS in the plasma of AIMD-BDL mice where elevated expression of inflammatory genes and decreased expression of hepatic detoxification enzymes were also found in liver when compared to the BDL group. These findings indicate that gut microbiota plays a critical role in cholestatic liver injury. Maintaining its homeostasis may alleviate liver injury in patients with cholestasis.
Collapse
Affiliation(s)
- Xueqian Zhou
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoxun Zhang
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Nan Zhao
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Liangjun Zhang
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wen Qiu
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chunwei Song
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jin Chai
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shiying Cai
- The Liver Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Wensheng Chen
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
26
|
Deng Y, Liu X, Yao Y, Xiao B, He C, Guo S, Tang S, Qu X. The potential role of palygorskite and probiotics complex on the laying performance and faecal microbial community in Xuefeng black-bone chicken. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2022.2149357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yuying Deng
- Hunan Engineering Research Center of Poultry Production Safety, Hunan Co-Innovation Center of Animal Production Safety, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xu Liu
- Hunan Key Laboratory for Conservation and Utilization of Biological Resources in the Nanyue Mountainous Region, College of Life Sciences, Hengyang Normal University, Hengyang, Hunan, China
| | - Yaling Yao
- Hunan Yunfeifeng Agricultural Co. Ltd, Huaihua, China
| | - Bing Xiao
- Hunan Yunfeifeng Agricultural Co. Ltd, Huaihua, China
| | - Changqing He
- Hunan Engineering Research Center of Poultry Production Safety, Hunan Co-Innovation Center of Animal Production Safety, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Songchang Guo
- Hunan Engineering Research Center of Poultry Production Safety, Hunan Co-Innovation Center of Animal Production Safety, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Shengguo Tang
- Hunan Engineering Research Center of Poultry Production Safety, Hunan Co-Innovation Center of Animal Production Safety, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiangyong Qu
- Hunan Engineering Research Center of Poultry Production Safety, Hunan Co-Innovation Center of Animal Production Safety, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| |
Collapse
|
27
|
Akuzum B, Lee JY. Context-Dependent Regulation of Type17 Immunity by Microbiota at the Intestinal Barrier. Immune Netw 2022; 22:e46. [PMID: 36627936 PMCID: PMC9807962 DOI: 10.4110/in.2022.22.e46] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 12/30/2022] Open
Abstract
T-helper-17 (Th17) cells and related IL-17-producing (type17) lymphocytes are abundant at the epithelial barrier. In response to bacterial and fungal infection, the signature cytokines IL-17A/F and IL-22 mediate the antimicrobial immune response and contribute to wound healing of injured tissues. Despite their protective function, type17 lymphocytes are also responsible for various chronic inflammatory disorders, including inflammatory bowel disease (IBD) and colitis associated cancer (CAC). A deeper understanding of type17 regulatory mechanisms could ultimately lead to the discovery of therapeutic strategies for the treatment of chronic inflammatory disorders and the prevention of cancer. In this review, we discuss the current understanding of the development and function of type17 immune cells at the intestinal barrier, focusing on the impact of microbiota-immune interactions on intestinal barrier homeostasis and disease etiology.
Collapse
Affiliation(s)
- Begum Akuzum
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
28
|
The Ameliorative Effect and Mechanisms of Ruditapes philippinarum Bioactive Peptides on Obesity and Hyperlipidemia Induced by a High-Fat Diet in Mice. Nutrients 2022; 14:nu14235066. [PMID: 36501096 PMCID: PMC9737393 DOI: 10.3390/nu14235066] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
In this study, bioactive peptides (RBPs) from Ruditapes philippinarum were prepared by fermentation with Bacillus natto and the effect and mechanisms of RBPs on obesity and hyperlipidemia were explored in mice. We found that RBPs significantly reduced body weight, adipose tissue weight, accumulation of hepatic lipids, and serum levels of total cholesterol (CHO), triglyceride (TG), and low-density lipoprotein (LDL). Mechanistic studies showed that RBPs up-regulated the hepatic expression of genes related to lipolysis, such as hormone-sensitive lipase (HSL), phosphorylated AMP-activated protein kinase (p-AMPK), and peroxisome proliferator-activated receptors α (PPARα), and down-regulated the expression of peroxisome proliferator-activated receptors γ (PPARγ) which is related to lipid synthesis. In addition, RBPs could attenuate obesity and hyperlipidemia by regulating disordered gut microbiota composition, such as increasing the abundance of microflora related to the synthesis of short chain fatty acids (SCFAs) (Bacteroidetes, Prevotellaceas_UCG_001, norank_f_Muribaculaceae, and Odoribacter) and controlling those related to intestinal inflammation (reduced abundance of Deferribacteres and increased abundance of Alistipes and ASF356) to exert anti-obesity and lipid-lowering activities. Our findings laid the foundation for the development and utilization of RBPs as a functional food to ameliorate obesity and hyperlipidemia.
Collapse
|
29
|
Yin L, Huang G, Khan I, Su L, Xia W, Law BYK, Wong VKW, Wu Q, Wang J, Leong WK, Hsiao WLW. Poria cocos polysaccharides exert prebiotic function to attenuate the adverse effects and improve the therapeutic outcome of 5-FU in Apc Min/+ mice. Chin Med 2022; 17:116. [PMID: 36192796 PMCID: PMC9531437 DOI: 10.1186/s13020-022-00667-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As a first-line chemotherapeutic agent, 5-fluorouracil (5-FU) exhibits many side effects, weakening its efficacy in cancer treatment. In this study, we hypothesize that Poria cocos polysaccharides (PCP), a traditional Chinese herbal medicine with various bioactivities and prebiotic effects, might improve the therapeutic effect of 5-FU by restoring the homeostasis of the gut microenvironment and the commensal gut microflora. METHODS ApcMin/+ mice were employed to evaluate the anti-cancer effect of 5-FU in conjunction with PCP treatment. Body weight and food consumption were monitored weekly. Polyp count was used to assess the anti-cancer effect of PCP and 5-FU. Expressions of mucosal cytokines and gut epithelial junction molecules were measured using qRT-PCR. 16S rRNA gene sequencing of fecal DNAs was used to evaluate the compositional changes of gut microbiota (GM). Transplantation of Lactobacillus johnsonii and Bifidobacterium animalis were performed to verify the prebiotic effects of PCP in improving the efficacy of 5-FU. RESULTS The results showed that PCP treatment alleviated the weight loss caused by 5-FU treatment and reduced the polyp burden in ApcMin/+ mice. Additionally, PCP treatment eased the cytotoxic effects of 5-FU by reducing the expressions of pro-inflammatory cytokines, increasing the anti-inflammatory cytokines; and significantly improving the gut barriers by enhancing the tight junction proteins and associated adhesion molecules. Furthermore, 16S rRNA gene sequencing data showed that PCP alone or with 5-FU could stimulate the growth of probiotic bacteria (Bacteroides acidifaciens, Bacteroides intestinihominis, Butyricicoccus pullicaecorum, and the genera Lactobacillus, Bifidobacterium, Eubacterium). At the same time, it inhibited the growth of potential pathogens (e.g., Alistipes finegoldii, Alistipes massiliensis, Alistipes putredinis., Citrobacter spp., Desulfovibrio spp., and Desulfovibrio desulfuricans). Moreover, the results showed that transplantation of L.johnsonii and B.animalis effectively reduced the polyp burden in ApcMin/+ mice being treated with 5-FU. CONCLUSION Our study showed that PCP could effectively improve the anti-cancer effect of 5-FU by attenuating its side effects, modulating intestinal inflammation, improving the gut epithelial barrier, and modulating the gut microbiota of ApcMin/+ mice.
Collapse
Affiliation(s)
- Lin Yin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Guoxin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China.,Clinical Research Center, Shantou Central Hospital, Shantou, China.,Zhuhai MUST Science and Technology Research Institute, Zhuhai, China
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Lu Su
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Wenrui Xia
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Qiang Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Jingyi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Wai Kit Leong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - W L Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China. .,Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China.
| |
Collapse
|
30
|
Jiang ZF, Wu W, Hu HB, Li ZY, Zhong M, Zhang L. P2X7 receptor as the regulator of T-cell function in intestinal barrier disruption. World J Gastroenterol 2022; 28:5265-5279. [PMID: 36185635 PMCID: PMC9521516 DOI: 10.3748/wjg.v28.i36.5265] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
The intestinal mucosa is a highly compartmentalized structure that forms a direct barrier between the host intestine and the environment, and its dysfunction could result in a serious disease. As T cells, which are important components of the mucosal immune system, interact with gut microbiota and maintain intestinal homeostasis, they may be involved in the process of intestinal barrier dysfunction. P2X7 receptor (P2X7R), a member of the P2X receptors family, mediates the effects of extracellular adenosine triphosphate and is expressed by most innate or adaptive immune cells, including T cells. Current evidence has demonstrated that P2X7R is involved in inflammation and mediates the survival and differentiation of T lymphocytes, indicating its potential role in the regulation of T cell function. In this review, we summarize the available research about the regulatory role and mechanism of P2X7R on the intestinal mucosa-derived T cells in the setting of intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Zhi-Feng Jiang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Wei Wu
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Han-Bing Hu
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Zheng-Yang Li
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
31
|
Wang S, Kang X, Alenius H, Wong SH, Karisola P, El-Nezami H. Oral exposure to Ag or TiO 2 nanoparticles perturbed gut transcriptome and microbiota in a mouse model of ulcerative colitis. Food Chem Toxicol 2022; 169:113368. [PMID: 36087619 DOI: 10.1016/j.fct.2022.113368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022]
Abstract
Silver (nAg) and titanium dioxide (nTiO2) nanoparticles improve texture, flavour or anti-microbial properties of various food products and packaging materials. Despite their increased oral exposure, their potential toxicities in the dysfunctional intestine are unclear. Here, the effects of ingested nAg or nTiO2 on inflamed colon were revealed in a mouse model of chemical-induced acute ulcerative colitis. Mice (eight/group) were exposed to nAg or nTiO2 by oral gavage for 10 consecutive days. We characterized disease phenotypes, histology, and alterations in colonic transcriptome (RNA sequencing) and gut microbiome (16S sequencing). Oral exposure to nAg caused only minor changes in phenotypic hallmarks of colitic mice but induced extensive responses in gene expression enriching processes of apoptotic cell death and RNA metabolism. Instead, ingested nTiO2 yielded shorter colon, aggravated epithelial hyperplasia and deeper infiltration of inflammatory cells. Both nanoparticles significantly changed the gut microbiota composition, resulting in loss of diversity and increase of potential pathobionts. They also increased colonic mucus and abundance of Akkermansia muciniphila. Overall, nAg and nTiO2 induce dissimilar immunotoxicological changes at the molecular and microbiome level in the context of colon inflammation. The results provide valuable information for evaluation of utilizing metallic nanoparticles in food products for the vulnerable population.
Collapse
Affiliation(s)
- Shuyuan Wang
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China.
| | - Xing Kang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Harri Alenius
- Human Microbiome Research Program, University of Helsinki, Haartmaninkatu 3, 00290, Helsinki, Finland; Institute of Environmental Medicine (IMM), Karolinska Institutet, Stockholm, 171 77, Sweden.
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| | - Piia Karisola
- Human Microbiome Research Program, University of Helsinki, Haartmaninkatu 3, 00290, Helsinki, Finland.
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China; Nutrition and Health, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
32
|
Li X, Wu X, Wang Q, Xu W, Zhao Q, Xu N, Hu X, Ye Z, Yu S, Liu J, He X, Shi F, Zhang Q, Li W. Sanguinarine ameliorates DSS induced ulcerative colitis by inhibiting NLRP3 inflammasome activation and modulating intestinal microbiota in C57BL/6 mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154321. [PMID: 35843190 DOI: 10.1016/j.phymed.2022.154321] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/27/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Sanguinarine (SAN) is an important natural anti-inflammatory constitutes and dietary supplementation with SAN could improve the relative length of the intestine, alter gut microbiota, and enhance growth performance of pigs, broiler chickens, and cattle. However, it is unclear whether it has the therapeutic effect on ulcerative colitis (UC). PURPOSE This study aimed to investigate the therapeutic effect of SAN on UC and explore its mechanisms of action. STUDY DESIGN AND METHODS Several efficacy indexes of SAN on dextran sulfate sodium (DSS)-induced C57BL/6 mice were evaluated. ELISA kit and western blot analysis were used to evaluate it's anti-inflammatory effect and the mechanism of action. 16S rDNA sequencing detection was used to determine the impact of SAN on gut microbiota. RESULTS SAN and Sulfasalazine could significantly improve the colon length, the weight loss, the symptoms and the pathological injury of colon in DSS-induced mice. Meanwhile, SAN could decrease the levels of pro-inflammatory cytokines (TNF-α, IFN-γ, IL-1β, IL-6, IL-13 and IL-18) and increase the levels of anti-inflammatory cytokines (IL-4 and IL-10) in colon, and suppress DSS-induced high expressions of NLRP3, caspase-1 and IL-1β. In addition, SAN (0.5, 1 μM) could inhibit the expression level of NLRP3 and the activation of caspase-1 and IL-1β in lipopolysaccharide-stimulated THP-1 cells in non-cytotoxic doses, which was similar to that of MCC950, a specific inhibitor of NLRP3 inflammasome activation. The abundance changes of many genera such as Muribaculaceae_unclassified, Escherichia-Shigella, Lachnospiraceae_NK4A136_group and Helicobacter were also closely related to the improvement of SAN on intestinal inflammatory response. CONCLUSION SAN exhibited therapeutic effect on DSS-induced colitis by blocking NLRP3-(Caspase-1)/IL-1β pathway and improving intestinal microbial dysbiosis. SAN might be developed to treat UC and other disorders associated with microbial dysbiosis.
Collapse
Affiliation(s)
- Xiaodong Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Xia Wu
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qi Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Weilv Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Qingwei Zhao
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Nana Xu
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Xingjiang Hu
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ziqi Ye
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Songxia Yu
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jian Liu
- Department of Intensive Care Unit, the First Affiliated Hospital College of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China
| | - Xuelin He
- Department of Nephrology, Beilun People's Hospital, Ningbo 315826, Zhejiang Province, China
| | - Fushan Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China.
| | - Qiao Zhang
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China.
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China.
| |
Collapse
|
33
|
Huang Z, Zhan M, Cheng G, Lin R, Zhai X, Zheng H, Wang Q, Yu Y, Xu Z. IHNV Infection Induces Strong Mucosal Immunity and Changes of Microbiota in Trout Intestine. Viruses 2022; 14:v14081838. [PMID: 36016461 PMCID: PMC9415333 DOI: 10.3390/v14081838] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
The fish intestinal mucosa is among the main sites through which environmental microorganisms interact with the host. Therefore, this tissue not only constitutes the first line of defense against pathogenic microorganisms but also plays a crucial role in commensal colonization. The interaction between the mucosal immune system, commensal microbiota, and viral pathogens has been extensively described in the mammalian intestine. However, very few studies have characterized these interactions in early vertebrates such as teleosts. In this study, rainbow trout (Oncorhynchus mykiss) was infected with infectious hematopoietic necrosis virus (IHNV) via a recently developed immersion method to explore the effects of viral infection on gut immunity and microbial community structure. IHNV successfully invaded the gut mucosa of trout, resulting in severe tissue damage, inflammation, and an increase in gut mucus. Moreover, viral infection triggered a strong innate and adaptive immune response in the gut, and RNA−seq analysis indicated that both antiviral and antibacterial immune pathways were induced, suggesting that the viral infection was accompanied by secondary bacterial infection. Furthermore, 16S rRNA sequencing also revealed that IHNV infection induced severe dysbiosis, which was characterized by large increases in the abundance of Bacteroidetes and pathobiont proliferation. Moreover, the fish that survived viral infection exhibited a reversal of tissue damage and inflammation, and their microbiome was restored to its pre−infection state. Our findings thus demonstrated that the relationships between the microbiota and gut immune system are highly sensitive to the physiological changes triggered by viral infection. Therefore, opportunistic bacterial infection must also be considered when developing strategies to control viral infection.
Collapse
Affiliation(s)
- Zhenyu Huang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Mengting Zhan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Gaofeng Cheng
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Ruiqi Lin
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xue Zhai
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Haiou Zheng
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingchao Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yongyao Yu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen Xu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Correspondence:
| |
Collapse
|
34
|
Bailey MJ, Holzhausen EA, Morgan ZEM, Naik N, Shaffer JP, Liang D, Chang HH, Sarnat J, Sun S, Berger PK, Schmidt KA, Lurmann F, Goran MI, Alderete TL. Postnatal exposure to ambient air pollutants is associated with the composition of the infant gut microbiota at 6-months of age. Gut Microbes 2022; 14:2105096. [PMID: 35968805 PMCID: PMC9466616 DOI: 10.1080/19490976.2022.2105096] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Epidemiological studies in adults have shown that exposure to ambient air pollution (AAP) is associated with the composition of the adult gut microbiome, but these relationships have not been examined in infancy. We aimed to determine if 6-month postnatal AAP exposure was associated with the infant gut microbiota at 6 months of age in a cohort of Latino mother-infant dyads from the Southern California Mother's Milk Study (n = 103). We estimated particulate matter (PM2.5 and PM10) and nitrogen dioxide (NO2) exposure from birth to 6-months based on residential address histories. We characterized the infant gut microbiota using 16S rRNA amplicon sequencing at 6-months of age. At 6-months, the gut microbiota was dominated by the phyla Bacteroidetes, Firmicutes, Proteobacteria, and Actinobacteria. Our results show that, after adjusting for important confounders, postnatal AAP exposure was associated with the composition of the gut microbiota. As an example, PM10 exposure was positively associated with Dialister, Dorea, Acinetobacter, and Campylobacter while PM2.5 was positively associated with Actinomyces. Further, exposure to PM10 and PM2.5 was inversely associated with Alistipes and NO2 exposure was positively associated with Actinomyces, Enterococcus, Clostridium, and Eubacterium. Several of these taxa have previously been linked with systemic inflammation, including the genera Dialister and Dorea. This study provides the first evidence of significant associations between exposure to AAP and the composition of the infant gut microbiota, which may have important implications for future infant health and development.
Collapse
Affiliation(s)
- Maximilian J. Bailey
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | | | | | - Noopur Naik
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Justin P. Shaffer
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Donghai Liang
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Howard H. Chang
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jeremy Sarnat
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Shan Sun
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Paige K. Berger
- Department of Pediatrics, The Saban Research Institute, Children’s Hospital of Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Kelsey A. Schmidt
- Department of Pediatrics, The Saban Research Institute, Children’s Hospital of Los Angeles, University of Southern California, Los Angeles, CA, USA
| | | | - Michael I. Goran
- Department of Pediatrics, The Saban Research Institute, Children’s Hospital of Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA,CONTACT Tanya L. Alderete Department of Integrative Physiology, University of Colorado, Boulder, CO80309, USA
| |
Collapse
|
35
|
Lin Q, Kuypers M, Liu Z, Copeland JK, Chan D, Robertson SJ, Kontogiannis J, Guttman DS, Banks EK, Philpott DJ, Mallevaey T. Invariant natural killer T cells minimally influence gut microbiota composition in mice. Gut Microbes 2022; 14:2104087. [PMID: 35912530 PMCID: PMC9348128 DOI: 10.1080/19490976.2022.2104087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Invariant Natural Killer T (iNKT) cells are unconventional T cells that respond to glycolipid antigens found in microbes in a CD1d-dependent manner. iNKT cells exert innate-like functions and produce copious amounts of cytokines, chemokines and cytotoxic molecules within only minutes of activation. As such, iNKT cells can fuel or dampen inflammation in a context-dependent manner. In addition, iNKT cells provide potent immunity against bacteria, viruses, parasites and fungi. Although microbiota-iNKT cell interactions are not well-characterized, mounting evidence suggests that microbiota colonization early in life impacts iNKT cell homeostasis and functions in disease. In this study, we showed that CD1d-/- and Vα14 Tg mice, which lack and have increased numbers of iNKT cells, respectively, had no significant alterations in gut microbiota composition compared to their littermate controls. Furthermore, specific iNKT cell activation by glycolipid antigens only resulted in a transient and minimal shift in microbiota composition when compared to the natural drift found in our colony. Our findings demonstrate that iNKT cells have little to no influence in regulating commensal bacteria at steady state.Abbreviations: iNKT: invariant Natural Killer T cell; αGC: α-galactosylceramide.
Collapse
Affiliation(s)
- Qiaochu Lin
- Department of Immunology, University of Toronto, Toronto, ON, Canada,CONTACT Thierry Mallevaey University of Toronto, Department of Immunology, Medical Sciences Building, Room 7334,1 King’s College Circle, Toronto, OntarioM5S 1A8, Canada
| | - Meggie Kuypers
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Zhewei Liu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Donny Chan
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Susan J Robertson
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jean Kontogiannis
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - E. Kate Banks
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, ON, Canada,Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
Lai S, Wang J, Wang B, Wang R, Li G, Jia Y, Chen T, Chen Y. Alterations in gut microbiota affect behavioral and inflammatory responses to methamphetamine in mice. Psychopharmacology (Berl) 2022; 239:1-16. [PMID: 35503371 DOI: 10.1007/s00213-022-06154-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 04/21/2022] [Indexed: 11/30/2022]
Abstract
RATIONALE AND OBJECTIVES Methamphetamine (METH) is a highly addictive and widely abused drug that causes severe neuroinflammation in the human brain. The gut microbiota has a tremendous impact on the core symptoms of neuropsychiatric disorders via the microbiota-gut-brain (MGB) axis. However, it is not clear whether alterations in the gut microbiota are involved in METH exposure. METHODS We established a mouse model with chronic, escalating doses of METH exposure. Intervene in gut microbiota with antibiotics to observe the changes of locomotor activity caused by METH exposure in mice. qPCR and 16S rRNA gene sequencing were used to analyze the gut microbiota profiles. In addition, we tested the levels of inflammatory factors in the nucleus accumbens (NAc), prefrontal cortex (mPFC), hippocampus (HIp), and spleen. Finally, short-chain fatty acids (SCFAs) were supplemented to determine the interaction between behavior changes and the structure of gut microbiota. RESULTS In this research, METH increased the locomotor activity of mice, while antibiotics changed the effect. Antibiotics enhanced the expression of pro-inflammatory cytokines in mPFC, HIp, and spleen of METH-exposed mice. METH altered the gut microbiota of mice after antibiotic treatment, such as Butyricicoccus and Roseburia, which are related to butyrate metabolism. Supplementation with SCFAs changed the behavior of METH-exposed mice and decreased Parabacteroides and increased Lactobacillus in METH-exposed mice gut. CONCLUSIONS This research showed that antibiotics affected the behavior of METH-exposed mice and promoted inflammation. Our findings suggest that SCFAs might regulate METH-induced gut microbiota changes and behavior.
Collapse
Affiliation(s)
- Simin Lai
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China
| | - Jing Wang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China
| | - Biao Wang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China
| | - Rui Wang
- Forensic Medicine College, Key Laboratory of the Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Guodong Li
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, the Second Affiliated Hospital of Xi'an Jiaotong University College of Medicin, Xi'an, People's Republic of China
| | - Yuwei Jia
- Department of Laboratory Medicine, Baoji Maternal and Child Health Hospital, Baoji, People's Republic of China
| | - Teng Chen
- Forensic Medicine College, Key Laboratory of the Health Ministry for Forensic Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, People's Republic of China.
| |
Collapse
|
37
|
Wang L, Dong XL, Qin XM, Li ZY. Investigating the inter-individual variability of Astragali Radix against cisplatin-induced liver injury via 16S rRNA gene sequencing and LC/MS-based metabolomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154107. [PMID: 35561503 DOI: 10.1016/j.phymed.2022.154107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 03/31/2022] [Accepted: 04/14/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Cisplatin (CDDP), one of the widely used chemotherapeutic drugs, can induce a series of side effects, such as hepatotoxicity and gastrointestinal toxicity. Astragali Radix (AR) is widely used as the tonic herbal medicine in traditional Chinese medicine (TCM). However, there was no report about the hepatoprotective effect of AR against the cisplatin-induced hepatic damage. PURPOSE This study aimed to investigate the protective effect and potential mechanism of AR water extract against the cisplatin-induced liver injury. METHODS Cisplatin was utilized to induce the liver injury using ICR mice, and the protective effect of AR was evaluated by serum biochemistry indices and liver histopathology. Then UHPLC Q-TOF-MS/MS-based untargeted serum metabolomics approach combined with 16S rRNA-based microbiota analysis was used to explore the underlying biomarkers and mechanism about the liver-protective effect of AR. RESULTS AR could decrease the serum AST and ALT, ameliorate hepatic pathological damages caused by cisplatin. Serum metabolomics indicated AR could regulate the biosynthesis of unsaturated fatty acids, arachidonic acid metabolism, purine metabolism, and fatty acid biosynthesis. In addition, 16S rRNA gene sequencing analysis showed that AR could regulate cisplatin-induced gut microbiota disorder, especially the inflammation-related bacteria (p_Deferribacteres, g_Enterococcus, and g_Alistipes, etc.), and the short chain fatty acids (SCFAs)-producing bacteria (g_Alloprevotella, g_Intestinimoas, and g_Flavonifractor). Moreover, 7 mice (AR-7) showed better liver protective effect than the other 3 mice (AR-3), and their regulatory effect on the gut microbiota and serum metabolites were also different, indicating the presence of inter-individual variability for the liver protective effect of AR. CONCLUSIONS This study revealed the protective effect and the potential mechanisms of AR against cisplatin-induced liver injury, and found that inter-individual variability of the liver protective effect of AR was related to the host microbiome and metabolome. These findings provided new insight into the health effect of dietary AR as a functional food for cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Ling Wang
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China
| | - Xian-Long Dong
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China
| | - Zhen-Yu Li
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China.
| |
Collapse
|
38
|
Kovács AD, Langin LM, Hernandez JLG, Pearce DA. Acidified drinking water attenuates motor deficits and brain pathology in a mouse model of a childhood neurodegenerative disorder. Sci Rep 2022; 12:9025. [PMID: 35637265 PMCID: PMC9151921 DOI: 10.1038/s41598-022-12981-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractWe recently demonstrated that HCl-acidified drinking water, which is widely used in laboratory animal facilities, had some beneficial effects in the Cln3−/− mouse model of juvenile Batten disease, a neurodegenerative lysosomal storage disorder1. Here we tested if acidified drinking water has therapeutic effects in Cln1R151X nonsense mutant mice, a model of the infantile form of Batten disease. In Cln1R151X mice, acidified drinking water received from weaning prevented the impairment in pole climbing ability measured at 3 and 6 months of age. Histopathological analysis of the brain at 6 months showed that acidified drinking water decreased the amount of lysosomal storage material, reduced astrocytosis in the striatum and somatosensory barrelfield cortex, and attenuated microglial activation in the thalamus. Compared to wild-type mice, the gut microbiota of Cln1R151X mice was markedly different. Acidified drinking water significantly altered the gut microbiota composition of Cln1R151X mice, indicating a contribution of gut bacteria to the therapeutic effects of acidified water. Our results in Cln1R151X mice suggest that acidified drinking water may have beneficial effects for patients with infantile Batten disease. This study also verifies that acidified drinking water can modify disease phenotypes in mouse models, contributing to the inter-laboratory variations in neurological and pathological findings.
Collapse
|
39
|
Mashaqi S, Kallamadi R, Matta A, Quan SF, Patel SI, Combs D, Estep L, Lee-Iannotti J, Smith C, Parthasarathy S, Gozal D. Obstructive Sleep Apnea as a Risk Factor for COVID-19 Severity-The Gut Microbiome as a Common Player Mediating Systemic Inflammation via Gut Barrier Dysfunction. Cells 2022; 11:1569. [PMID: 35563874 PMCID: PMC9101605 DOI: 10.3390/cells11091569] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023] Open
Abstract
The novel corona virus that is now known as (SARS-CoV-2) has killed more than six million people worldwide. The disease presentation varies from mild respiratory symptoms to acute respiratory distress syndrome and ultimately death. Several risk factors have been shown to worsen the severity of COVID-19 outcomes (such as age, hypertension, diabetes mellitus, and obesity). Since many of these risk factors are known to be influenced by obstructive sleep apnea, this raises the possibility that OSA might be an independent risk factor for COVID-19 severity. A shift in the gut microbiota has been proposed to contribute to outcomes in both COVID-19 and OSA. To further evaluate the potential triangular interrelationships between these three elements, we conducted a thorough literature review attempting to elucidate these interactions. From this review, it is concluded that OSA may be a risk factor for worse COVID-19 clinical outcomes, and the shifts in gut microbiota associated with both COVID-19 and OSA may mediate processes leading to bacterial translocation via a defective gut barrier which can then foster systemic inflammation. Thus, targeting biomarkers of intestinal tight junction dysfunction in conjunction with restoring gut dysbiosis may provide novel avenues for both risk detection and adjuvant therapy.
Collapse
Affiliation(s)
- Saif Mashaqi
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, AZ 85719, USA; (S.F.Q.); (S.I.P.); (D.C.); (L.E.); (S.P.)
| | - Rekha Kallamadi
- Department of Internal Medicine, The University of North Dakota School of Medicine, Grand Forks, ND 58203, USA; (R.K.); (A.M.)
| | - Abhishek Matta
- Department of Internal Medicine, The University of North Dakota School of Medicine, Grand Forks, ND 58203, USA; (R.K.); (A.M.)
| | - Stuart F. Quan
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, AZ 85719, USA; (S.F.Q.); (S.I.P.); (D.C.); (L.E.); (S.P.)
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Salma I. Patel
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, AZ 85719, USA; (S.F.Q.); (S.I.P.); (D.C.); (L.E.); (S.P.)
| | - Daniel Combs
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, AZ 85719, USA; (S.F.Q.); (S.I.P.); (D.C.); (L.E.); (S.P.)
| | - Lauren Estep
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, AZ 85719, USA; (S.F.Q.); (S.I.P.); (D.C.); (L.E.); (S.P.)
| | - Joyce Lee-Iannotti
- Department of Sleep Medicine, The University of Arizona College of Medicine, Phoenix, AZ 85006, USA;
| | - Charles Smith
- The Intermountain Healthcare, Merrill Gappmayer Family Medicine Center, Provo, UT 84604, USA;
| | - Sairam Parthasarathy
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, AZ 85719, USA; (S.F.Q.); (S.I.P.); (D.C.); (L.E.); (S.P.)
| | - David Gozal
- Department of Child Health, University of Missouri School of Medicine, Columbia, MO 65201, USA;
| |
Collapse
|
40
|
Pre-Administration of Berberine Exerts Chemopreventive Effects in AOM/DSS-Induced Colitis-Associated Carcinogenesis Mice via Modulating Inflammation and Intestinal Microbiota. Nutrients 2022; 14:nu14040726. [PMID: 35215376 PMCID: PMC8879943 DOI: 10.3390/nu14040726] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
Inflammatory activation and intestinal flora imbalance play an essential role in the development and progression of colorectal cancer (CRC). Berberine (BBR) has attracted great attention in recent years due to its heath-related benefits in inflammatory disorders and tumors, but the intricate mechanisms have not been fully elucidated. In this study, the effects and the mechanism of BBR on colon cancer were investigated in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis-associated carcinogenesis mice model. Our results showed that pre-administration of BBR showed a decrease in weight loss, disease activity index (DAI) score, and the number of colon tumors in mice, compared with the model group. The evidence from pathological examination indicated that the malignancy of intestinal tumors was ameliorated after pre-administration of BBR. Additionally, pre-administration with BBR suppressed the expression of pro-inflammatory factors (interleukin (IL)-6, IL-1β, cyclooxygenase (COX)-2 and tumor necrosis factor (TNF)-α) and the cell-proliferation marker Ki67, while expression of the tight junction proteins (ZO-1 and occludin) were increased in colon tissue. Moreover, the levels of critical pathway proteins involved in the inflammatory process (p-STAT3 and p-JNK) and cell cycle regulation molecules (β-catenin, c-Myc and CylinD1) exhibited lower expression levels. Besides, 16S rRNA sequence analysis indicated that pre-administration of BBR increased the ratio of Firmicutes/Bacteroidetes (F:M) and the relative abundance of potentially beneficial bacteria, while the abundance of cancer-related bacteria was decreased. Gavage with Lactobacillus rhamnosus can improve the anti-tumor effect of BBR. Overall, pre-administration of BBR exerts preventive effects in colon carcinogenesis, and the mechanisms underlying these effects are correlated with the inhibition of inflammation and tumor proliferation and the maintenance of intestinal homeostasis.
Collapse
|
41
|
Shi Y, Zou Y, Xiong Y, Zhang S, Song M, An X, Liu C, Zhang W, Chen S. Host Gasdermin D restrains systemic endotoxemia by capturing Proteobacteria in the colon of high-fat diet-feeding mice. Gut Microbes 2021; 13:1946369. [PMID: 34275417 PMCID: PMC8288038 DOI: 10.1080/19490976.2021.1946369] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gasdermin D (GSDMD) functions as a key pyroptotic executor through its secreted N-terminal domain (GSDMD-N). However, the functional relevance and mechanistic basis of the precise roles of host colonic GSDMD in high-fat diet (HFD)-induced gut dysbiosis and systemic endotoxemia remain elusive. In this study, we demonstrate that HFD feeding triggers GSDMD-N secretion of both T-lymphocytes and enterocytes in mouse colons. GSDMD deficiency aggravates HFD-induced systemic endotoxemia, gut barrier impairment, and colonic inflammation. More importantly, active GSDMD-N kills the Proteobacteria phylum via directly interacting with Cardiolipin. Mechanistically, we identify that the Glu236 (a known residue for GSDMD protein cleavage) is a bona fide important site for the bacterial recognition of GSDMD. Collectively, our findings explain the mechanism by which colonic GSDMD-N maintains low levels of HFD-induced metabolic endotoxemia. A GSDMD-N mimetic containing an exposed Glu236 site could be an attractive strategy for the treatment of HFD-induced metabolic endotoxemia.
Collapse
Affiliation(s)
- Yujie Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yixin Zou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yonghong Xiong
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shiyao Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,Wenxiang Zhang State Key Laboratory of Natural Medicines, China Pharmaceutical University, #639 Longmian Avenue, Nanjing211198, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China,School of Life Science and Technology, China Pharmaceutical University, Nanjing, China,CONTACT Siyu Chen
| |
Collapse
|
42
|
Baghdadi MB, Ayyaz A, Coquenlorge S, Chu B, Kumar S, Streutker C, Wrana JL, Kim TH. Enteric glial cell heterogeneity regulates intestinal stem cell niches. Cell Stem Cell 2021; 29:86-100.e6. [PMID: 34727519 DOI: 10.1016/j.stem.2021.10.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/09/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022]
Abstract
The high turnover and regenerative capacity of the adult intestine relies on resident stem cells located at the bottom of the crypt. The enteric nervous system consists of an abundant network of enteric glial cells (EGCs) and neurons. Despite the close proximity of EGCs to stem cells, their in vivo role as a stem cell niche is still unclear. By analyzing the mouse and human intestinal mucosa transcriptomes at the single-cell level, we defined the regulation of EGC heterogeneity in homeostasis and chronic inflammatory bowel disease. Ablation of EGC subpopulations revealed that the repair potential of intestinal stem cells (ISCs) is regulated by a specific subset of glial fibrillary acidic protein (GFAP)+ EGCs. Mechanistically, injury induces expansion of GFAP+ EGCs, which express several WNT ligands to promote LGR5+ ISC self-renewal. Our work reveals the dynamically regulated heterogeneity of EGCs as a key part of the intestinal stem cell niche in regeneration and disease.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Arshad Ayyaz
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Sabrina Coquenlorge
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Bonnie Chu
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sandeep Kumar
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Catherine Streutker
- Department of Laboratory Medicine, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Jeffrey L Wrana
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
43
|
α-Galactosylceramide-Reactive NKT Cells Increase IgG1 Class Switch against a Clostridioides difficile Polysaccharide Antigen and Enhance Immunity against a Live Pathogen Challenge. Infect Immun 2021; 89:e0043821. [PMID: 34424751 DOI: 10.1128/iai.00438-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
All clinical Clostridioides difficile strains identified to date express a surface capsule-like polysaccharide structure known as polysaccharide II (PSII). The PSII antigen is immunogenic and, when conjugated to a protein carrier, induces a protective antibody response in animal models. Given that CD1d-restricted natural killer T (NKT) cells promote antibody responses, including those against carbohydrates, we tested the hypothesis that immunization with PSII and a CD1d-binding glycolipid adjuvant could lead to enhanced protection against a live C. difficile challenge. We purified PSII from a clinical isolate of C. difficile and immunized B6 mice with PSII alone or PSII plus the CD1d-binding glycolipid α-galactosylceramide (α-GC). PSII-specific IgM and IgG titers were evident in sera from immunized mice. The inclusion of α-GC had a modest influence on isotype switch but increased the IgG1/IgG2c ratio. Enhanced protection against C. difficile disease was achieved by inclusion of the α-GC ligand and was associated with reduced bacterial numbers in fecal pellets. In contrast, NKT-deficient Traj18-/- mice were not protected by the PSII/α-GC immunization modality. Absence of NKT cells similarly had a modest effect on isotype switch, but ratios of IgG1/IgG2c decreased. These results indicate that α-GC-driven NKT cells move the humoral immune response against C. difficile PSII antigen toward Th2-driven IgG1 and may contribute to augmented protection. This study suggests that NKT activation represents a pathway for additional B-cell help that could be used to supplement existing efforts to develop vaccines against polysaccharides derived from C. difficile and other pathogens.
Collapse
|
44
|
Abokor AA, McDaniel GH, Golonka RM, Campbell C, Brahmandam S, Yeoh BS, Joe B, Vijay-Kumar M, Saha P. Immunoglobulin A, an Active Liaison for Host-Microbiota Homeostasis. Microorganisms 2021; 9:2117. [PMID: 34683438 PMCID: PMC8539215 DOI: 10.3390/microorganisms9102117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Mucosal surfaces in the gastrointestinal tract are continually exposed to native, commensal antigens and susceptible to foreign, infectious antigens. Immunoglobulin A (IgA) provides dual humoral responses that create a symbiotic environment for the resident gut microbiota and prevent the invasion of enteric pathogens. This review features recent immunological and microbial studies that elucidate the underlying IgA and microbiota-dependent mechanisms for mutualism at physiological conditions. IgA derailment and concurrent microbiota instability in pathological diseases are also discussed in detail. Highlights of this review underscore that the source of IgA and its structural form can dictate microbiota reactivity to sustain a diverse niche where both host and bacteria benefit. Other important studies emphasize IgA insufficiency can result in the bloom of opportunistic pathogens that encroach the intestinal epithelia and disseminate into circulation. The continual growth of knowledge in these subjects can lead to the development of therapeutics targeting IgA and/or the microbiota to treat life threatening diseases.
Collapse
Affiliation(s)
- Ahmed A. Abokor
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Grant H. McDaniel
- College of Medicine, University of Toledo, Toledo, OH 43614, USA; (G.H.M.); (C.C.); (S.B.)
| | - Rachel M. Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Connor Campbell
- College of Medicine, University of Toledo, Toledo, OH 43614, USA; (G.H.M.); (C.C.); (S.B.)
| | - Sreya Brahmandam
- College of Medicine, University of Toledo, Toledo, OH 43614, USA; (G.H.M.); (C.C.); (S.B.)
| | - Beng San Yeoh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Bina Joe
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| |
Collapse
|
45
|
Early development of the skin microbiome: therapeutic opportunities. Pediatr Res 2021; 90:731-737. [PMID: 32919387 PMCID: PMC7952468 DOI: 10.1038/s41390-020-01146-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/23/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023]
Abstract
As human skin hosts a diverse microbiota in health and disease, there is an emerging consensus that dysregulated interactions between host and microbiome may contribute to chronic inflammatory disease of the skin. Neonatal skin is a unique habitat, structurally similar to the adult but with a different profile of metabolic substrates, environmental stressors, and immune activity. The surface is colonized within moments of birth with a bias toward maternal strains. Initial colonists are outcompeted as environmental exposures increase and host skin matures. Nonetheless, early life microbial acquisitions may have long-lasting effects on health through modulation of host immunity and competitive interactions between bacteria. Microbial ecology and its influence on health have been of interest to dermatologists for >50 years, and an explosion of recent interest in the microbiome has prompted ongoing investigations of several microbial therapeutics for dermatological disease. In this review, we consider how recent insight into the host and microbial factors driving development of the skin microbiome in early life offers new opportunities for therapeutic intervention. IMPACT: Advancement in understanding molecular mechanisms of bacterial competition opens new avenues of investigation into dermatological disease. Primary development of the skin microbiome is determined by immunological features of the cutaneous habitat. Understanding coordinated microbial and immunological development in the pediatric patient requires a multidisciplinary synthesis of primary literature.
Collapse
|
46
|
Morales Fénero C, Amaral MA, Xavier IK, Padovani BN, Paredes LC, Takiishi T, Lopes-Ferreira M, Lima C, Colombo A, Saraiva Câmara NO. Short chain fatty acids (SCFAs) improves TNBS-induced colitis in zebrafish. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:142-154. [PMID: 35492385 PMCID: PMC9040093 DOI: 10.1016/j.crimmu.2021.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022] Open
Abstract
The short-chain fatty acids (SCFAs) are metabolites originated from the fermentation of dietary fibers and amino acids produced by the bacteria of the intestinal microbiota. The most abundant SCFAs, acetate, propionate, and butyrate, have been proposed as a treatment for inflammatory bowel diseases (IBDs) due to their anti-inflammatory properties. This work aimed to analyze the effects of the treatment of three combined SCFAs in TNBS-induced intestinal inflammation in zebrafish larvae. Here, we demonstrated that SCFAs significantly increased the survival of TNBS-exposed larvae, preserved the intestinal endocytic function, reduced the expression of inflammatory cytokines and the intestinal recruitment of neutrophils caused by TNBS. However, SCFAs treatment did not appear to avoid TNBS-induced tissue damage in the intestinal wall and did not restore the number of mucus-producing goblet cells. Finally, exposure to TNBS induced dysbiosis of the microbiota with an increase in Betaproteobacteria and Actinobacteria, while the treatment with SCFAs maintained these population levels similar to control. Thus, we demonstrate that the treatment of three combined SCFAs presented anti-inflammatory properties previously seen in mammals, opening an opportunity to use zebrafish to explore the potential benefit of these and other metabolites to treat inflammation.
Collapse
Affiliation(s)
- Camila Morales Fénero
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Izabella Karina Xavier
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Barbara Nunes Padovani
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lais Cavalieri Paredes
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tatiana Takiishi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Mônica Lopes-Ferreira
- Center of Toxins, Immune Response and Cellular Signalling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Carla Lima
- Center of Toxins, Immune Response and Cellular Signalling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Alicia Colombo
- Department of Pathologic Anatomy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Nephrology Division, Federal University of São Paulo, Brazil
| |
Collapse
|
47
|
Vomhof-DeKrey EE, Stover A, Basson MD. Microbiome diversity declines while distinct expansions of Th17, iNKT, and dendritic cell subpopulations emerge after anastomosis surgery. Gut Pathog 2021; 13:51. [PMID: 34376235 PMCID: PMC8353768 DOI: 10.1186/s13099-021-00447-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/30/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Anastomotic failure causes morbidity and mortality even in technically correct anastomoses. Initial leaks must be prevented by mucosal reapproximation across the anastomosis. Healing is a concerted effort between intestinal epithelial cells (IECs), immune cells, and commensal bacteria. IEC TLR4 activation and signaling is required for mucosal healing, leading to inflammatory factor release that recruits immune cells to limit bacteria invasion. TLR4 absence leads to mucosal damage from loss in epithelial proliferation, attenuated inflammatory response, and bacteria translocation. We hypothesize after anastomosis, an imbalance in microbiota will occur due to a decrease in TLR4 expression and will lead to changes in the immune milieu. RESULTS We isolated fecal content and small intestinal leukocytes from murine, Roux-en-Y and end-to-end anastomoses, to identify microbiome changes and subsequent alterations in the regulatory and pro-inflammatory immune cells 3 days post-operative. TLR4+ IECs were impaired after anastomosis. Microbiome diversity was reduced, with Firmicutes, Bacteroidetes, and Saccharibacteria decreased and Proteobacteria increased. A distinct TCRβhi CD4+ T cells subset after anastomosis was 10-20-fold greater than in control mice. 84% were Th17 IL-17A/F+ IL-22+ and/or TNFα+. iNKT cells were increased and TCRβhi. 75% were iNKT IL-10+ and 13% iNKTh17 IL-22+. Additionally, Treg IL-10+ and IL-22+ cells were increased. A novel dendritic cell subset was identified in anastomotic regions that was CD11bhi CD103mid and was 93% IL-10+. CONCLUSIONS This anastomotic study demonstrated a decrease in IEC TLR4 expression and microbiome diversity which then coincided with increased expansion of regulatory and pro-inflammatory immune cells and cytokines. Defining the anastomotic mucosal environment could help inform innovative therapeutics to target excessive pro-inflammatory invasion and microbiome imbalance.
Collapse
Affiliation(s)
- Emilie E. Vomhof-DeKrey
- Department of Surgery, University of North Dakota School of Medicine and the Health Sciences, 1301 North Columbia Road, Stop 9037, Grand Forks, ND 58202 USA
- Department of Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, 1301 North Columbia Road, Stop 9037, Grand Forks, ND 58202 USA
| | - Allie Stover
- Department of Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, 1301 North Columbia Road, Stop 9037, Grand Forks, ND 58202 USA
| | - Marc D. Basson
- Department of Surgery, University of North Dakota School of Medicine and the Health Sciences, 1301 North Columbia Road, Stop 9037, Grand Forks, ND 58202 USA
- Department of Biomedical Sciences, University of North Dakota School of Medicine and the Health Sciences, 1301 North Columbia Road, Stop 9037, Grand Forks, ND 58202 USA
- Department of Pathology, University of North Dakota School of Medicine and the Health Sciences, 1301 North Columbia Road, Stop 9037, Grand Forks, ND 58202 USA
| |
Collapse
|
48
|
Herp S, Durai Raj AC, Salvado Silva M, Woelfel S, Stecher B. The human symbiont Mucispirillum schaedleri: causality in health and disease. Med Microbiol Immunol 2021; 210:173-179. [PMID: 34021796 PMCID: PMC7615636 DOI: 10.1007/s00430-021-00702-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/26/2022]
Abstract
Trillions of bacteria inhabit the mammalian gastrointestinal tract. In the majority of hosts, these symbionts contribute largely to beneficial functions promoting microbe-host homeostasis. However, an increasing number of human diseases is associated with altered microbiota composition and enrichment of certain bacterial species. A well-known example of this is Mucispirillum schaedleri, which has been associated with inflammatory conditions in the intestine. Mucispirillum spp. belong to the phylum Deferribacteres and are prevalent but low abundant members of the rodent, pig and human microbiota. Recently, M. schaedleri was causally linked to the development of Crohn's disease-like colitis in immunodeficient mice. While this study certifies a considerable pathogenic potential, the same organism can also promote health in the immunocompetent host: M. schaedleri protects from Salmonella enterica serovar Typhimurium (S. Tm)-induced colitis by interfering with the expression of the pathogen´s invasion machinery. In this review, we summarize the current knowledge on the mammalian gut symbiont M. schaedleri and its role in intestinal homeostasis and discuss open questions and perspectives for future research.
Collapse
Affiliation(s)
- Simone Herp
- Max-Von-Pettenkofer Institute, LMU Munich, Pettenkoferstr. 9a, Munich, Germany.
| | | | - Marta Salvado Silva
- Max-Von-Pettenkofer Institute, LMU Munich, Pettenkoferstr. 9a, Munich, Germany
| | - Simon Woelfel
- Max-Von-Pettenkofer Institute, LMU Munich, Pettenkoferstr. 9a, Munich, Germany
| | - Bärbel Stecher
- Max-Von-Pettenkofer Institute, LMU Munich, Pettenkoferstr. 9a, Munich, Germany
| |
Collapse
|
49
|
Regulation and Functions of Protumoral Unconventional T Cells in Solid Tumors. Cancers (Basel) 2021; 13:cancers13143578. [PMID: 34298791 PMCID: PMC8304984 DOI: 10.3390/cancers13143578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/02/2021] [Accepted: 07/12/2021] [Indexed: 01/03/2023] Open
Abstract
The vast majority of studies on T cell biology in tumor immunity have focused on peptide-reactive conventional T cells that are restricted to polymorphic major histocompatibility complex molecules. However, emerging evidence indicated that unconventional T cells, including γδ T cells, natural killer T (NKT) cells and mucosal-associated invariant T (MAIT) cells are also involved in tumor immunity. Unconventional T cells span the innate-adaptive continuum and possess the unique ability to rapidly react to nonpeptide antigens via their conserved T cell receptors (TCRs) and/or to activating cytokines to orchestrate many aspects of the immune response. Since unconventional T cell lineages comprise discrete functional subsets, they can mediate both anti- and protumoral activities. Here, we review the current understanding of the functions and regulatory mechanisms of protumoral unconventional T cell subsets in the tumor environment. We also discuss the therapeutic potential of these deleterious subsets in solid cancers and why further feasibility studies are warranted.
Collapse
|
50
|
Mohamed SS, Abdeltawab NF, Wadie W, Ahmed LA, Ammar RM, Rabini S, Abdel-Aziz H, Khayyal MT. Effect of the standard herbal preparation, STW5, treatment on dysbiosis induced by dextran sodium sulfate in experimental colitis. BMC Complement Med Ther 2021; 21:168. [PMID: 34103031 PMCID: PMC8188707 DOI: 10.1186/s12906-021-03337-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/05/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The standardized herbal preparation, STW 5, is effective clinically in functional gastrointestinal disorders and experimentally in ulcerative colitis (UC). The present study explores whether the beneficial effect of STW 5 involves influencing the intestinal microbiota. METHODS UC was induced in Wistar rats by feeding them 5% dextran sodium sulfate (DSS) in drinking water for 7 days. Rats were treated concurrently with STW 5 and sacrificed 24 h after last drug administration. Fecal samples were used to determine changes in the abundance of selected microbial phyla and genera using real-time PCR. RESULTS Induction of UC led to dysbiosis and changes in the gut microbiota. The changes included an increase in some genera of the Firmicutes, namely Enterococcus, and a decrease in others, namely Blautia, Clostridium, and Lactobacillus. DSS further induced a marked increase in the abundance of Bacteroidetes and Proteobacteria as well as in the relative abundance of Actinobacteria and its genus Bifidobacterium. Methanobrevibacter levels (phylum Euryarchaeota) were also increased. Microbial dysbiosis was associated with changes in various parameters of colonic inflammation. STW 5 effectively guarded against those changes and significantly affected the indices of edema and inflammation in the UC model. Changes in colon length, colon mass index, inflammatory and apoptotic markers, and histological changes induced by DSS were also prevented. CONCLUSIONS Dysbiosis plays a contributing role in the development of DSS-induced UC. Derangements in the microbial flora and associated inflammatory processes were largely prevented by STW 5, suggesting that this effect might contribute towards its beneficial usefulness in this condition.
Collapse
Affiliation(s)
- Sarah S Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Nourtan F Abdeltawab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Ramy M Ammar
- Bayer Consumer Health, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
- Pharmacology Department, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Sabine Rabini
- Bayer Consumer Health, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Heba Abdel-Aziz
- Bayer Consumer Health, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Mohamed T Khayyal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| |
Collapse
|