1
|
Wilson BK, Romanova S, Bronich TK, Prud'homme RK. Intestinal distribution of anionic, cationic, and neutral polymer-stabilized nanocarriers measured with a lanthanide (europium) tracer assay. J Control Release 2024; 376:200-214. [PMID: 39374745 DOI: 10.1016/j.jconrel.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Nanocarriers, more commonly called nanoparticles (NPs), have found increasing use as delivery vehicles which increase the oral bioavailability of poorly water-soluble and peptide therapeutics. Therapeutic bioavailability is commonly assessed by measuring plasma concentrations that reflect the absorption kinetics. This bioavailability is a convolution of the gastrointestinal distribution of the NP vehicle, the release rate of the encapsulated therapeutic cargo, and the absorption-metabolism-distribution kinetics of the released therapeutic. The spatiotemporal distribution of the NP vehicle in the gastrointestinal tract is not well studied and is a buried parameter in PK studies used to measure the effectiveness of an NP formulation. This work is a study of the intestinal distribution and fate of orally dosed NPs in male CD-1 mice over 24 h. NPs have identical hydrophobic cores - composed of poly(styrene) homopolymer, a naphthalocyanine dye, and oleate-coated europium oxide colloids - with one of four different surface stabilizers: neutral poly(styrene)-block-poly(ethylene glycol) (PS-b-PEG), moderately negative hydroxypropyl methylcellulose acetate succinate (HPMCAS), highly negative poly(styrene)-block-poly(acrylic acid) (PS-b-PAA), and highly cationic adsorbed chitosan HCl on PS-b-PAA stabilized NPs. NP hydrodynamic diameters are all below 200 nm, with some variation attributable to the molecular properties of the stabilizing polymer. The encapsulated hydrophobic europium oxide colloids do not release soluble europium ions, enabling the use of highly sensitive inductively coupled plasma mass spectrometry (ICP-MS) to detect NP concentrations in digested biological tissues. Highly anionically-charged PAA and cationically-charged chitosan stabilized NPs showed statistically significant increased retention compared to the neutral PEG-stabilized NPs at p < 0.05 significance and (1-β) > 0.95 power. HPMCAS-stabilized NPs showed statistically insignificant greater retention than PEG-stabilized NPs, and all NP formulations showed clearance from the intestines within 24 h. Different surface charges preferentially reside in different segments of the intestines, where cationic chitosan-stabilized NPs showed increased retention in the small intestines (ileum) and anionic PAA-stabilized NPs in the large intestines (caecum and colon). Modifying the surface charge of a NP can be used to modulate mucoadhesion, total retention, and intestinal segment specific retention, which enables the rational design of delivery vehicles that maximize residence times in appropriate locations.
Collapse
Affiliation(s)
- Brian K Wilson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA
| | - Svetlana Romanova
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Robert K Prud'homme
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
2
|
Demirturk M, Cinar MS, Avci FY. The immune interactions of gut glycans and microbiota in health and disease. Mol Microbiol 2024; 122:313-330. [PMID: 38703041 DOI: 10.1111/mmi.15267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
The human digestive system harbors a vast diversity of commensal bacteria and maintains a symbiotic relationship with them. However, imbalances in the gut microbiota accompany various diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancers (CRCs), which significantly impact the well-being of populations globally. Glycosylation of the mucus layer is a crucial factor that plays a critical role in maintaining the homeostatic environment in the gut. This review delves into how the gut microbiota, immune cells, and gut mucus layer work together to establish a balanced gut environment. Specifically, the role of glycosylation in regulating immune cell responses and mucus metabolism in this process is examined.
Collapse
Affiliation(s)
- Mahmut Demirturk
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mukaddes Sena Cinar
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fikri Y Avci
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Zhong K, Chen X, Zhang J, Jiang X, Zhang J, Huang M, Bi S, Ju C, Luo Y. Recent Advances in Oral Vaccines for Animals. Vet Sci 2024; 11:353. [PMID: 39195807 PMCID: PMC11360704 DOI: 10.3390/vetsci11080353] [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: 07/01/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Compared to traditional injected vaccines, oral vaccines offer significant advantages for the immunization of livestock and wildlife due to their ease of use, high compliance, improved safety, and potential to stimulate mucosal immune responses and induce systemic immunity against pathogens. This review provides an overview of the delivery methods for oral vaccines, and the factors that influence their immunogenicity. We also highlight the global progress and achievements in the development and use of oral vaccines for animals, shedding light on potential future applications in this field.
Collapse
Affiliation(s)
- Kaining Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Xinting Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Junhao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Xiaoyu Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Junhui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Minyi Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
| | - Shuilian Bi
- School of Food Science, Guangdong Pharmaceutical University, Zhongshan 528458, China;
| | - Chunmei Ju
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
- Key Laboratory of Animal Vaccine Development of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510640, China
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510640, China; (K.Z.); (X.C.); (J.Z.); (X.J.); (J.Z.); (M.H.)
- Key Laboratory of Animal Vaccine Development of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510640, China
| |
Collapse
|
4
|
Asgari R, Bazzazan MA, Karimi Jirandehi A, Yousefzadeh S, Alaei M, Keshavarz Shahbaz S. Peyer's Patch: Possible target for modulating the Gut-Brain-Axis through microbiota. Cell Immunol 2024; 401-402:104844. [PMID: 38901288 DOI: 10.1016/j.cellimm.2024.104844] [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: 04/04/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The gastrointestinal (GI) tract and the brain form bidirectional nervous, immune, and endocrine communications known as the gut-brain axis. Several factors can affect this axis; among them, various studies have focused on the microbiota and imply that alterations in microbiota combinations can influence both the brain and GI. Also, many studies have shown that the immune system has a vital role in varying gut microbiota combinations. In the current paper, we will review the multidirectional effects of gut microbiota, immune system, and nervous system on each other. Specifically, this review mainly focuses on the impact of Peyer's patches as a critical component of the gut immune system on the gut-brain axis through affecting the gut's microbial composition. In this way, some factors were discussed as proposed elements of missing gaps in this field.
Collapse
Affiliation(s)
- Reza Asgari
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Mohammad Amin Bazzazan
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Ashkan Karimi Jirandehi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Salar Yousefzadeh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Masood Alaei
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Sanaz Keshavarz Shahbaz
- USERN Office, Qazvin University of Medical science, Qazvin, Iran; Cellular and Molecular Research Center, Research Institute for prevention of Non- Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
5
|
Lawrence DA, Jadhav A, Mondal TK, Carson K, Lee WT, Hogan AH, Herbst KW, Michelow IC, Brimacombe M, Salazar JC. Inflammatory and Autoimmune Aspects of Multisystem Inflammatory Syndrome in Children (MIS-C): A Prospective Cohort Study. Viruses 2024; 16:950. [PMID: 38932242 PMCID: PMC11209514 DOI: 10.3390/v16060950] [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: 04/30/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Multisystem Inflammatory Syndrome in Children (MIS-C) is a potentially life-threatening complication of COVID-19. The pathophysiological mechanisms leading to severe disease are poorly understood. This study leveraged clinical samples from a well-characterized cohort of children hospitalized with COVID-19 or MIS-C to compare immune-mediated biomarkers. Our objective was to identify selected immune molecules that could explain, in part, why certain SARS-CoV-2-infected children developed MIS-C. We hypothesized that type-2 helper T cell-mediated inflammation can elicit autoantibodies, which may account for some of the differences observed between the moderate-severe COVID-19 (COVID+) and MIS-C cohort. We enumerated blood leukocytes and measured levels of selected serum cytokines, chemokines, antibodies to COVID-19 antigens, and autoantibodies in children presenting to an academic medical center in Connecticut, United States. The neutrophil/lymphocyte and eosinophil/lymphocyte ratios were significantly higher in those in the MIS-C versus COVID+ cohort. IgM and IgA, but not IgG antibodies to SARS-CoV-2 receptor binding domain were significantly higher in the MIS-C cohort than the COVID+ cohort. The serum levels of certain type-2 cytokines (interleukin (IL)-4, IL-5, IL-6, IL-8, IL-10, IL-13, and IL-33) were significantly higher in children with MIS-C compared to the COVID+ and SARS-CoV-2-negative cohorts. IgG autoantibodies to brain antigens and pentraxin were higher in children with MIS-C compared to SARS-CoV-19-negative controls, and children with MIS-C had higher levels of IgG anti-contactin-associated protein-like 2 (caspr2) compared to the COVID+ and SARS-CoV-19-negative controls. We speculate that autoimmune responses in certain COVID-19 patients may induce pathophysiological changes that lead to MIS-C. The triggers of autoimmunity and factors accounting for type-2 inflammation require further investigation.
Collapse
Affiliation(s)
- David A. Lawrence
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (A.J.); (T.K.M.); (K.C.); (W.T.L.)
- School of Public Health, University at Albany, Rensselaer, NY 12144, USA
| | - Aishwarya Jadhav
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (A.J.); (T.K.M.); (K.C.); (W.T.L.)
| | - Tapan K. Mondal
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (A.J.); (T.K.M.); (K.C.); (W.T.L.)
| | - Kyle Carson
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (A.J.); (T.K.M.); (K.C.); (W.T.L.)
| | - William T. Lee
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA; (A.J.); (T.K.M.); (K.C.); (W.T.L.)
- School of Public Health, University at Albany, Rensselaer, NY 12144, USA
| | - Alexander H. Hogan
- Division of Hospital Medicine, Connecticut Children’s, Hartford, CT 06106, USA;
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA; (I.C.M.); (M.B.); (J.C.S.)
| | - Katherine W. Herbst
- Division of Pediatric Infectious Diseases and Immunology, Connecticut Children’s, Hartford, CT 06106, USA;
- Department of Research, Connecticut Children’s Research Institute, Hartford, CT 06106, USA
| | - Ian C. Michelow
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA; (I.C.M.); (M.B.); (J.C.S.)
- Division of Pediatric Infectious Diseases and Immunology, Connecticut Children’s, Hartford, CT 06106, USA;
| | - Michael Brimacombe
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA; (I.C.M.); (M.B.); (J.C.S.)
- Department of Research, Connecticut Children’s Research Institute, Hartford, CT 06106, USA
| | - Juan C. Salazar
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030, USA; (I.C.M.); (M.B.); (J.C.S.)
- Division of Pediatric Infectious Diseases and Immunology, Connecticut Children’s, Hartford, CT 06106, USA;
| | | |
Collapse
|
6
|
Hong C, Huang Y, Yang G, Wen X, Wang L, Yang X, Gao K, Jiang Z, Xiao H. Maternal resveratrol improves the intestinal health and weight gain of suckling piglets during high summer temperatures: The involvement of exosome-derived microRNAs and immunoglobin in colostrum. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:36-48. [PMID: 38464951 PMCID: PMC10921242 DOI: 10.1016/j.aninu.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/17/2023] [Accepted: 01/18/2024] [Indexed: 03/12/2024]
Abstract
Previous studies have shown that maternal resveratrol improved growth performance and altered the microbial composition of suckling piglets under hot summer conditions. However, it remains unclear how maternal resveratrol improves growth performance of suckling piglets during high summer temperatures. A total of 20 sows (Landrace × Large White; three parity) were randomly assigned to 2 groups (with or without 300 mg/kg resveratrol) from d 75 of gestation to d 21 of lactation during high ambient temperatures (from 27 to 30 °C). The results showed that maternal resveratrol supplementation increased total daily weight gain of piglets under hot summer conditions, which is consistent with previous studies. Furthermore, we found that maternal resveratrol improved the intestinal morphology and intestinal epithelial proliferation in suckling piglets. Dietary resveratrol supplementation affected the characteristics of exosome-derived microRNAs (miRNAs) in sow colostrum, as well as the genes targeted by differentially produced miRNAs. MiRNAs are concentrated in the tight junction pathway. As a result, the expression of intestinal tight junction proteins was increased in suckling piglets (P < 0.05). Notably, maternal resveratrol increased the intestinal secretory immunoglobulin A (sIgA) levels of suckling piglets via colostrum immunoglobin (P < 0.05), which could increase the abundance of beneficial microbiota to further increase the concentration of short chain fatty acids (SCFA) in suckling piglets' intestine (P < 0.05). Finally, our correlation analysis further demonstrated the positive associations between significantly differential intestinal microbiota, intestinal sIgA production and SCFA concentrations, as well as the positive relation between total daily weight gain and intestinal health of suckling piglets. Taken together, our findings suggested that maternal resveratrol could promote intestinal health to improve piglet growth during high summer temperatures, which might be associated with the immunoglobin and exosome-derived miRNAs in sows' colostrum.
Collapse
Affiliation(s)
- Changming Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Institute of Subtropical Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yujian Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Guan Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Xiaolu Wen
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Xuefen Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Kaiguo Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Zongyong Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Hao Xiao
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| |
Collapse
|
7
|
Dishaw LJ, Litman GW, Liberti A. Tethering of soluble immune effectors to mucin and chitin reflects a convergent and dynamic role in gut immunity. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230078. [PMID: 38497268 PMCID: PMC10945408 DOI: 10.1098/rstb.2023.0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/04/2023] [Indexed: 03/19/2024] Open
Abstract
The immune system employs soluble effectors to shape luminal spaces. Antibodies are soluble molecules that effect immunological responses, including neutralization, opsonization, antibody-dependent cytotoxicity and complement activation. These molecules are comprised of immunoglobulin (Ig) domains. The N-terminal Ig domains recognize antigen, and the C-terminal domains facilitate their elimination through phagocytosis (opsonization). A less-recognized function mediated by the C-terminal Ig domains of the IgG class of antibodies (Fc region) involves the formation of multiple low-affinity bonds with the mucus matrix. This association anchors the antibody molecule to the matrix to entrap potential pathogens. Even though invertebrates are not known to have antibodies, protochordates have a class of secreted molecules containing Ig domains that can bind bacteria and potentially serve a similar purpose. The VCBPs (V region-containing chitin-binding proteins) possess a C-terminal chitin-binding domain that helps tether them to chitin-rich mucus gels, mimicking the IgG-mediated Fc trapping of microbes in mucus. The broad functional similarity of these structurally divergent, Ig-containing, secreted effectors makes a case for a unique form of convergent evolution within chordates. This opinion essay highlights emerging evidence that divergent secreted immune effectors with Ig-like domains evolved to manage immune recognition at mucosal surfaces in strikingly similar ways. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- L. J. Dishaw
- Morsani College of Medicine, Department of Pediatrics, University of South Florida, Children's Research Institute, St. Petersburg, FL 33701, USA
| | - G. W. Litman
- Morsani College of Medicine, Department of Pediatrics, University of South Florida, Children's Research Institute, St. Petersburg, FL 33701, USA
| | - A. Liberti
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, 80122 Naples, Italy
| |
Collapse
|
8
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|
9
|
Ashonibare VJ, Akorede BA, Ashonibare PJ, Akhigbe TM, Akhigbe RE. Gut microbiota-gonadal axis: the impact of gut microbiota on reproductive functions. Front Immunol 2024; 15:1346035. [PMID: 38482009 PMCID: PMC10933031 DOI: 10.3389/fimmu.2024.1346035] [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: 11/28/2023] [Accepted: 01/30/2024] [Indexed: 04/12/2024] Open
Abstract
The influence of gut microbiota on physiological processes is rapidly gaining attention globally. Despite being under-studied, there are available data demonstrating a gut microbiota-gonadal cross-talk, and the importance of this axis in reproduction. This study reviews the impacts of gut microbiota on reproduction. In addition, the possible mechanisms by which gut microbiota modulates male and female reproduction are presented. Databases, including Embase, Google scholar, Pubmed/Medline, Scopus, and Web of Science, were explored using relevant key words. Findings showed that gut microbiota promotes gonadal functions by modulating the circulating levels of steroid sex hormones, insulin sensitivity, immune system, and gonadal microbiota. Gut microbiota also alters ROS generation and the activation of cytokine accumulation. In conclusion, available data demonstrate the existence of a gut microbiota-gonadal axis, and role of this axis on gonadal functions. However, majority of the data were compelling evidences from animal studies with a great dearth of human data. Therefore, human studies validating the reports of experimental studies using animal models are important.
Collapse
Affiliation(s)
- Victory J. Ashonibare
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Bolaji A. Akorede
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Biomedical Sciences, University of Wyoming, Laramie, WY, United States
| | - Precious J. Ashonibare
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Tunmise M. Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Breeding and Genetic Unit, Department of Agronomy, Osun State University, Ejigbo, Osun State, Nigeria
| | - Roland Eghoghosoa Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
10
|
Hajra D, Kirthivasan N, Chakravortty D. Symbiotic Synergy from Sponges to Humans: Microflora-Host Harmony Is Crucial for Ensuring Survival and Shielding against Invading Pathogens. ACS Infect Dis 2024; 10:317-336. [PMID: 38170903 DOI: 10.1021/acsinfecdis.3c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Gut microbiota plays several roles in the host organism's metabolism and physiology. This phenomenon holds across different species from different kingdoms and classes. Different species across various classes engage in continuous crosstalk via various mechanisms with their gut microbiota, ensuring homeostasis of the host. In this Review, the diversity of the microflora, the development of the microflora in the host, its regulations by the host, and its functional implications on the host, especially in the context of dysbiosis, are discussed across different organisms from sponges to humans. Overall, our review aims to address the indispensable nature of the microbiome in the host's survival, fitness, and protection against invading pathogens.
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore, Karnataka-560012, India
| | - Nikhita Kirthivasan
- Undergraduate Programme, Indian Institute of Science, Bangalore, Karnataka-560012, India
| | - Dipshikha Chakravortty
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore, Karnataka-560012, India
| |
Collapse
|
11
|
Abstract
Biogeography is the study of species distribution and diversity within an ecosystem and is at the core of how we understand ecosystem dynamics and interactions at the macroscale. In gut microbial communities, a historical reliance on bulk sequencing to probe community composition and dynamics has overlooked critical processes whereby microscale interactions affect systems-level microbiota function and the relationship with the host. In recent years, higher-resolution sequencing and novel single-cell level data have uncovered an incredible heterogeneity in microbial composition and have enabled a more nuanced spatial understanding of the gut microbiota. In an era when spatial transcriptomics and single-cell imaging and analysis have become key tools in mammalian cell and tissue biology, many of these techniques are now being applied to the microbiota. This fresh approach to intestinal biogeography has given important insights that span temporal and spatial scales, from the discovery of mucus encapsulation of the microbiota to the quantification of bacterial species throughout the gut. In this Review, we highlight emerging knowledge surrounding gut biogeography enabled by the observation and quantification of heterogeneity across multiple scales.
Collapse
Affiliation(s)
- Giselle McCallum
- Department of Biology, Concordia University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carolina Tropini
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
- Humans and the Microbiome Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Eriksen C, Moll JM, Myers PN, Pinto ARA, Danneskiold-Samsøe NB, Dehli RI, Rosholm LB, Dalgaard MD, Penders J, Jonkers DM, Pan-Hammarström Q, Hammarström L, Kristiansen K, Brix S. IgG and IgM cooperate in coating of intestinal bacteria in IgA deficiency. Nat Commun 2023; 14:8124. [PMID: 38065985 PMCID: PMC10709418 DOI: 10.1038/s41467-023-44007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Immunoglobulin A (IgA) is acknowledged to play a role in the defence of the mucosal barrier by coating microorganisms. Surprisingly, IgA-deficient humans exhibit few infection-related complications, raising the question if the more specific IgG may help IgM in compensating for the lack of IgA. Here we employ a cohort of IgA-deficient humans, each paired with IgA-sufficient household members, to investigate multi-Ig bacterial coating. In IgA-deficient humans, IgM alone, and together with IgG, recapitulate coating of most bacterial families, despite an overall 3.6-fold lower Ig-coating. Bacterial IgG coating is dominated by IgG1 and IgG4. Single-IgG2 bacterial coating is sparse and linked to enhanced Escherichia coli load and TNF-α. Although single-IgG2 coating is 1.6-fold more prevalent in IgA deficiency than in healthy controls, it is 2-fold less prevalent than in inflammatory bowel disease. Altogether we demonstrate that IgG assists IgM in coating of most bacterial families in the absence of IgA and identify single-IgG2 bacterial coating as an inflammatory marker.
Collapse
Affiliation(s)
- Carsten Eriksen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
| | - Janne Marie Moll
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pernille Neve Myers
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ana Rosa Almeida Pinto
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Rasmus Ibsen Dehli
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Lisbeth Buus Rosholm
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - John Penders
- Department of Medical Microbiology, Infectious Diseases and Infection Prevention, NUTRIM School for Nutrition and Translational Research in Metabolism & Care and Public Health Research Institute CAPHRI, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Daisy Mae Jonkers
- Division Gastroenterology-Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translation Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Qiang Pan-Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Lennart Hammarström
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karsten Kristiansen
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- BGI-Shenzhen, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, Qingdao, Shandong, China
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark.
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark.
| |
Collapse
|
13
|
Qin D, Li Y, Chen X, Li L, Wang G, Hou X, Yu L. Secretory IgA-ETEC F5 Immune Complexes Promote Dendritic Cell Differentiation and Prime T Cell Proliferation in the Mouse Intestine. Life (Basel) 2023; 13:1936. [PMID: 37763339 PMCID: PMC10532461 DOI: 10.3390/life13091936] [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: 08/23/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Although secretory IgA (SIgA) is the dominant antibody in mucosal secretions, the capacity of the SIgA-antigen complex to prime the activation of dendritic cells (DCs) and T cells in the intestinal epithelium is not well understood. To this end, the SIgA-ETEC F5 immune complexes (ICs) were prepared via Ni-NTA pull-down. After injecting the ICs into the intestines of SPF BALB/c mice, most ICs were observed in the Peyer's patch (PP). We established a microfold (M) cell culture model in vitro for transport experiments and the inhibition test. To evaluate the priming effect of mucosal immunity, we employed the DC2.4 stimulation test, T lymphocyte proliferation assays, and cytokine detection assays. We found that the ICs were taken up via clathrin-dependent endocytosis through M cells. The high expression of costimulatory molecules CD86, CD80, and CD40 indicated that the ICs promoted the differentiation and maturation of DC2.4 cells. The stimulation index (SI) in the complex group was significantly higher than in the control group, suggesting that the ICs stimulated the proliferation of primed T cells. The secretion of some cytokines, namely TNF-α, IFN-γ, IL-2, IL-4, IL-5, and IL-6, in spleen cells from the immunized mice was upregulated. These results indicate that ETEC F5 delivery mediated by SIgA in PPs initiates mucosal immune responses.
Collapse
Affiliation(s)
- Da Qin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (D.Q.); (Y.L.); (X.C.); (L.L.); (G.W.)
| | - Ying Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (D.Q.); (Y.L.); (X.C.); (L.L.); (G.W.)
| | - Xiaoyan Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (D.Q.); (Y.L.); (X.C.); (L.L.); (G.W.)
| | - Liyang Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (D.Q.); (Y.L.); (X.C.); (L.L.); (G.W.)
| | - Guihua Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (D.Q.); (Y.L.); (X.C.); (L.L.); (G.W.)
| | - Xilin Hou
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China;
| | - Liyun Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (D.Q.); (Y.L.); (X.C.); (L.L.); (G.W.)
| |
Collapse
|
14
|
Liu N, Zou S, Xie C, Meng Y, Xu X. Effect of the β-glucan from Lentinus edodes on colitis-associated colorectal cancer and gut microbiota. Carbohydr Polym 2023; 316:121069. [PMID: 37321711 DOI: 10.1016/j.carbpol.2023.121069] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
Colorectal cancer is the third most common cancer in the world, and therapies with safety are in great need. In this study, the β-glucan isolated from Lentinus edodes was successfully fractionated into three fractions with different weight-average molecular weight (Mw) by ultrasonic degradation and used for the treatment of colorectal cancer. In our findings, the β-glucan was successfully degraded with the Mw decreased from 2.56 × 106 Da to 1.41 × 106 Da, exhibiting the triple helix structure without conformation disruption. The in vitro results indicate that β-glucan fractions inhibited colon cancer cell proliferation, induced colon cancer cell apoptosis, and reduced inflammation. The in vivo results based on Azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model demonstrate that the lower-molecular weight β-glucan fraction showed stronger anti-inflammatory and anti-colon cancer activities by reconstructing intestinal mucosal barrier, increasing short chain fatty acids (SCFAs) content, regulating metabolism of gut microbiota, and rebuilding the gut microbiota structure with the increased Bacteroides and the decreased Proteobacteria at the phylum level, as well as with the decreased Helicobacter and the increased Muribaculum at the genus level. These findings provide scientific basis for using the β-glucan to regulate gut microbiota as an alternative strategy in the clinical treatment of colon cancer.
Collapse
Affiliation(s)
- Ningyue Liu
- College of Chemistry and Molecular Sciences, Hubei Engineering Center of Natural Polymers-based Medical Materials, Wuhan University, Wuhan 430072, China
| | - Siwei Zou
- College of Chemistry and Molecular Sciences, Hubei Engineering Center of Natural Polymers-based Medical Materials, Wuhan University, Wuhan 430072, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yan Meng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiaojuan Xu
- College of Chemistry and Molecular Sciences, Hubei Engineering Center of Natural Polymers-based Medical Materials, Wuhan University, Wuhan 430072, China; Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
15
|
Inaba R, Vujakovic S, Bergstrom K. The gut mucus network: A dynamic liaison between microbes and the immune system. Semin Immunol 2023; 69:101807. [PMID: 37478802 DOI: 10.1016/j.smim.2023.101807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 06/24/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
A complex mucus network made up of large polymers of the mucin-family glycoprotein MUC2 exists between the large intestinal microbial mass and epithelial and immune cells. This has long been understood as an innate immune defense barrier against the microbiota and other luminal threats that reinforces the barrier function of the epithelium and limits microbiota contact with the tissues. However, past and recent studies have provided new evidence of how critical the mucus network is to act as a 'liaison' between host and microbe to mediate anti-inflammatory, mutualistic interactions with the microbiota and protection from pathogens. This review summarizes historical and recent insights into the formation of the gut mucus network, how the microbes and immune system influence mucus, and in turn, how the mucus influences immune responses to the microbiota.
Collapse
Affiliation(s)
- Rain Inaba
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Sara Vujakovic
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Kirk Bergstrom
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada.
| |
Collapse
|
16
|
Gaglio SC, Perduca M, Zipeto D, Bardi G. Efficiency of Chitosan Nanocarriers in Vaccinology for Mucosal Immunization. Vaccines (Basel) 2023; 11:1333. [PMID: 37631901 PMCID: PMC10459455 DOI: 10.3390/vaccines11081333] [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: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023] Open
Abstract
The mucosal barrier constitutes a huge surface area, close to 40 m2 in humans, located mostly in the respiratory, gastrointestinal and urogenital tracts and ocular cavities. It plays a crucial role in tissue interactions with the microbiome, dietary antigens and other environmental materials. Effective vaccinations to achieve highly protective mucosal immunity are evolving strategies to counteract several serious diseases including tuberculosis, diphtheria, influenzae B, severe acute respiratory syndrome, Human Papilloma Virus infection and Acquired Immune Deficiency Syndrome. Interestingly, one of the reasons behind the rapid spread of severe acute respiratory syndrome coronavirus 2 variants has been the weakness of local immunization at the level of the respiratory mucosa. Mucosal vaccines can outperform parenteral vaccination as they specifically elicit protective mucosal immune responses blocking infection and transmission. In this scenario, chitosan-based nanovaccines are promising adjuvants-carrier systems that rely on the ability of chitosan to cross tight junctions and enhance particle uptake due to chitosan-specific mucoadhesive properties. Indeed, chitosan not only improves the adhesion of antigens to the mucosa promoting their absorption but also shows intrinsic immunostimulant abilities. Furthermore, by finely tuning the colloidal properties of chitosan, it can provide sustained antigen release to strongly activate the humoral defense. In the present review, we agnostically discuss the potential reasons why chitosan-based vaccine carriers, that efficiently elicit strong immune responses in experimental setups and in some pre-clinical/clinical studies, are still poorly considered for therapeutic formulations.
Collapse
Affiliation(s)
- Salvatore Calogero Gaglio
- Department of Biotechnology, University of Verona, Cà Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Massimiliano Perduca
- Department of Biotechnology, University of Verona, Cà Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Giuseppe Bardi
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
17
|
Mohammed AD, Ball RAW, Kubinak JL. The interplay between bile acids and mucosal adaptive immunity. PLoS Pathog 2023; 19:e1011356. [PMID: 37347728 PMCID: PMC10286976 DOI: 10.1371/journal.ppat.1011356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023] Open
Affiliation(s)
- Ahmed Dawood Mohammed
- Department of Pathology, Microbiology, Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Ryan A. W. Ball
- Department of Pathology, Microbiology, Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Jason L. Kubinak
- Department of Pathology, Microbiology, Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| |
Collapse
|
18
|
Yue Z, Fan Z, Zhang H, Feng B, Wu C, Chen S, Ouyang J, Fan H, Weng P, Feng H, Chen S, Dong M, Xu A, Huang S. Differential roles of the fish chitinous membrane in gut barrier immunity and digestive compartments. EMBO Rep 2023; 24:e56645. [PMID: 36852962 PMCID: PMC10074124 DOI: 10.15252/embr.202256645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 03/01/2023] Open
Abstract
The chitin-based peritrophic matrix (PM) is a structure critical for both gut immunity and digestion in invertebrates. PM was traditionally considered lost in all vertebrates, but a PM-like chitinous membrane (CM) has recently been discovered in fishes, which may increase the knowledge on vertebrate gut physiology and structural evolution. Here, we show that in zebrafish, the CM affects ingestion behavior, microbial homeostasis, epithelial renewal, digestion, growth, and longevity. Young mutant fish without CM appear healthy and are able to complete their life cycle normally, but with increasing age they develop gut inflammation, resulting in gut atrophy. Unlike mammals, zebrafish have no visible gel-forming mucin layers to protect their gut epithelia, but at least in young fish, the CM is not a prerequisite for the antibacterial gut immunity. These findings provide new insights into the role of the CM in fish prosperity and its eventual loss in tetrapods. These findings may also help to improve fish health and conservation, as well as to advance the understanding of vertebrate gut physiology and human intestinal diseases.
Collapse
Affiliation(s)
- Zirui Yue
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Zhaoyu Fan
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Hao Zhang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Buhan Feng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Chengyi Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | - Shenghui Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Jihua Ouyang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Huiping Fan
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Panwei Weng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Huixiong Feng
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Shangwu Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Meiling Dong
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
| | - Anlong Xu
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Shengfeng Huang
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangdongChina#
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
19
|
Song C, Chai Z, Chen S, Zhang H, Zhang X, Zhou Y. Intestinal mucus components and secretion mechanisms: what we do and do not know. Exp Mol Med 2023; 55:681-691. [PMID: 37009791 PMCID: PMC10167328 DOI: 10.1038/s12276-023-00960-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/26/2022] [Indexed: 04/04/2023] Open
Abstract
Damage to the colon mucus barrier, the first line of defense against microorganisms, is an important determinant of intestinal diseases such as inflammatory bowel disease and colorectal cancer, and disorder in extraintestinal organs. The mucus layer has attracted the attention of the scientific community in recent years, and with the discovery of new mucosal components, it has become increasingly clear that the mucosal barrier is a complex system composed of many components. Moreover, certain components are jointly involved in regulating the structure and function of the mucus barrier. Therefore, a comprehensive and systematic understanding of the functional components of the mucus layer is clearly warranted. In this review, we summarize the various functional components of the mucus layer identified thus far and describe their unique roles in shaping mucosal structure and function. Furthermore, we detail the mechanisms underlying mucus secretion, including baseline and stimulated secretion. In our opinion, baseline secretion can be categorized into spontaneous Ca2+ oscillation-mediated slow and continuous secretion and stimulated secretion, which is mediated by massive Ca2+ influx induced by exogenous stimuli. This review extends the current understanding of the intestinal mucus barrier, with an emphasis on host defense strategies based on fortification of the mucus layer.
Collapse
Affiliation(s)
- Chunyan Song
- Department of Preventive Medicine, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Zhenglong Chai
- Department of Preventive Medicine, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Si Chen
- Department of Preventive Medicine, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Hui Zhang
- Department of Preventive Medicine, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China
| | - Xiaohong Zhang
- Department of Preventive Medicine, Health Science Center, Ningbo University, Zhejiang Key Laboratory of Pathophysiology, Ningbo, Zhejiang, 315211, China.
- The Affiliated Hospital of Medical School, Ningbo University, Institute of Digestive Disease of Ningbo University, Ningbo, Zhejiang, 315020, China.
| | - Yuping Zhou
- The Affiliated Hospital of Medical School, Ningbo University, Institute of Digestive Disease of Ningbo University, Ningbo, Zhejiang, 315020, China.
| |
Collapse
|
20
|
Strachan E, Clemente-Casares X, Tsai S. Maternal provisions in type 1 diabetes: Evidence for both protective & pathogenic potential. Front Immunol 2023; 14:1146082. [PMID: 37033940 PMCID: PMC10073710 DOI: 10.3389/fimmu.2023.1146082] [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: 01/16/2023] [Accepted: 03/08/2023] [Indexed: 04/11/2023] Open
Abstract
Maternal influences on the immune health and development of an infant begin in utero and continue well into the postnatal period, shaping and educating the child's maturing immune system. Two maternal provisions include early microbial colonizers to initiate microbiota establishment and the transfer of antibodies from mother to baby. Maternal antibodies are a result of a lifetime of antigenic experience, reflecting the infection history, health and environmental exposure of the mother. These same factors are strong influencers of the microbiota, inexorably linking the two. Together, these provisions help to educate the developing neonatal immune system and shape lymphocyte repertoires, establishing a role for external environmental influences even before birth. In the context of autoimmunity, the transfer of maternal autoantibodies has the potential to be harmful for the child, sometimes targeting tissues and cells with devastating consequences. Curiously, this does not seem to apply to maternal autoantibody transfer in type 1 diabetes (T1D). Moreover, despite the rising prevalence of the disease, little research has been conducted on the effects of maternal dysbiosis or antibody transfer from an affected mother to her offspring and thus their relevance to disease development in the offspring remains unclear. This review seeks to provide a thorough evaluation of the role of maternal microorganisms and antibodies within the context of T1D, exploring both their pathogenic and protective potential. Although a definitive understanding of their significance in infant T1D development remains elusive at present, we endeavor to present what has been learned with the goal of spurring further interest in this important and intriguing question.
Collapse
Affiliation(s)
| | | | - Sue Tsai
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
21
|
Matsuzawa M, Ando T, Fukase S, Kimura M, Kume Y, Ide T, Izawa K, Kaitani A, Hara M, Nakamura E, Kamei A, Matsuda A, Nakano N, Maeda K, Tada N, Ogawa H, Okumura K, Murakami A, Ebihara N, Kitaura J. The protective role of conjunctival goblet cell mucin sialylation. Nat Commun 2023; 14:1417. [PMID: 36932081 PMCID: PMC10023771 DOI: 10.1038/s41467-023-37101-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
Gel-forming mucins secreted by conjunctival goblet cells have been implicated in the clearance of allergens, pathogens, and debris. However, their roles remain incompletely understood. Here we show that human and mouse conjunctival goblet cell mucins have Alcian blue-detectable sialic acids, but not sulfates in the steady state. Interestingly, Balb/c mouse strain lacks this sialylation due to a point mutation in a sialyltransferase gene, St6galnac1, which is responsible for sialyl-Tn synthesis. Introduction of intact St6galnac1 to Balb/c restores the sialylation of conjunctival goblet cell mucus. Sialylated mucus efficiently captures and encapsulates the allergen particles in an impenetrable layer, leading to the protection of mice from the development of allergic conjunctivitis. Expression of ST6GALNAC1 and sialyl-Tn is upregulated in humans under conditions with chronic stimuli. These results indicate that the sialylated glycans on the ocular mucins play an essential role in maintaining the conjunctival mucosa by protecting from the incoming foreign bodies such as allergen particles.
Collapse
Affiliation(s)
- Moe Matsuzawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
| | - Saaya Fukase
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Meiko Kimura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Yasuharu Kume
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Takuma Ide
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Otorhinolaryngology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Kumi Izawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Ayako Kaitani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Mutsuko Hara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Laboratory of Molecular and Biochemical Research, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Eri Nakamura
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Anna Kamei
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Akira Matsuda
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Nobuhiro Nakano
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Keiko Maeda
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
- Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Norihiro Tada
- Center for Biomedical Research Resources, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Nobuyuki Ebihara
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Urayasu, Chiba, 279-0021, Japan
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Jiro Kitaura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan.
| |
Collapse
|
22
|
He X, Chen X, Wang H, Du G, Sun X. Recent advances in respiratory immunization: A focus on COVID-19 vaccines. J Control Release 2023; 355:655-674. [PMID: 36787821 PMCID: PMC9937028 DOI: 10.1016/j.jconrel.2023.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
The development of vaccines has always been an essential task worldwide since vaccines are regarded as powerful weapons in protecting the global population. Although the vast majority of currently authorized human vaccinations are administered intramuscularly or subcutaneously, exploring novel routes of immunization has been a prominent area of study in recent years. This is particularly relevant in the face of pandemic diseases, such as COVID-19, where respiratory immunization offers distinct advantages, such as inducing systemic and mucosal responses to prevent viral infections in both the upper and lower respiratory tracts and also leading to higher patient compliance. However, the development of respiratory vaccines confronts challenges due to the physiological barriers of the respiratory tract, with most of these vaccines still in the research and development stage. In this review, we detail the structure of the respiratory tract and the mechanisms of mucosal immunity, as well as the obstacles to respiratory vaccination. We also examine the considerations necessary in constructing a COVID-19 respiratory vaccine, including the dosage form of the vaccines, potential excipients and mucosal adjuvants, and delivery systems and devices for respiratory vaccines. Finally, we present a comprehensive overview of the COVID-19 respiratory vaccines currently under clinical investigation. We hope this review can provide valuable insights and inspiration for the future development of respiratory vaccinations.
Collapse
Affiliation(s)
- Xiyue He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaoyan Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hairui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
23
|
Yang D, Hu X, Li H, Xu W, Wu T, Chen J. Molecular cloning and characteristic analysis of polymeric immunoglobulin receptor-like (plgRL) in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2023; 132:108503. [PMID: 36581255 DOI: 10.1016/j.fsi.2022.108503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
In the present study, the polyimmunoglobulin receptor-like (pIgRL) of large yellow croaker (Larimichthys crocea) was first cloned and characterized. LcpIgRL's full-length cDNA was 1610 bp, encoding 377 amino acids, and the protein's predicted molecular weight was 41.9 kDa, containing two immunoglobulin-like structural domains. The transcript levels of LcpIgRL in different tissues of healthy large yellow croaker were examined by real-time fluorescence quantitative PCR, and the results showed that the gills and head kidney had the highest levels. Within 36 h of the large yellow croaker being infected with Vibrio harveyi, pIgRL mRNA first increased and then decreased in all determined tissues, with the highest expression in the skin and hindgut. Furthermore, a recombinant protein of the extracellular region of LcpIgRL was expressed in E. coli BL21, and a murine rLcpIgRL polyclonal antibody was prepared, which could react specifically with the natural LcpIgRL in skin mucus, but no natural LcpIgRL was detected in serum. Meanwhile, it was found that the rLcpIgRL could bind to the recombinant IgM and the natural IgM, indicating that LcpIgRL could mediate the transport of IgM in mucus. In addition, rLcpIgRL binds to Aeromonas hydrophila and V. harveyi, as well as lipopolysaccharide (LPS) and various saccharides, and reduced binding to bacteria was observed under LPS treatment, suggesting that LcpIgRL can bind to bacteria to prevent infection and that saccharide binding is an important mechanism of interaction between pIgRL and bacteria.
Collapse
Affiliation(s)
- Du Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiaoman Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Hao Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Wenlong Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Ting Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
24
|
Qi C, Tu H, Zhao Y, Zhou J, Chen J, Hu H, Yu R, Sun J. Breast Milk-Derived Limosilactobacillus reuteri Prevents Atopic Dermatitis in Mice via Activating Retinol Absorption and Metabolism in Peyer's Patches. Mol Nutr Food Res 2023; 67:e2200444. [PMID: 36480309 DOI: 10.1002/mnfr.202200444] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/24/2022] [Indexed: 12/13/2022]
Abstract
SCOPE Supplementing Limosilactobacillus reuteri Fn041, a breast milk-derived probiotic from agricultural and pastoral areas, to maternal mice during late pregnancy and lactation prevents atopic dermatitis (AD) in offspring. This study aims to elucidate the molecular mechanism of Fn041-mediated immune regulation. METHODS AND RESULTS Fn041 is administered prenatal and postnatal to maternal mice, and to offspring after weaning. The ears are administered with calcipotriol to induce AD. Fn041 treatment significantly alleviates ear inflammation, and reduces mast cell infiltration. Fn041 treatment upregulates and downregulates intestinal ZO-1 and Claudin-2 mRNA expression, respectively. Transcriptome analysis of Peyer's patches reveals that pathways related to DNA damage repair are activated in AD mice, which is inhibited by Fn041 treatment. Fn041 activates pathways related to retinol absorption and metabolism. Untargeted metabolomic analysis reveals that Fn041 treatment increases plasma retinol and kynurenine. Fn041 treatment does not significantly alter the overall cecal microbiota profile, only increases the relative abundances of Ligilactobacillus apodemi, Ligilactobacillus murinus, Akkermansia muciniphila, and Bacteroides thetaiotaomicron. CONCLUSIONS Fn041 induces anti-AD immune responses directly by promoting the absorption and metabolism of retinol in Peyer's patches, and plays an indirect role by strengthening the mucosal barrier and increasing the abundance of specific anti-AD bacteria in the cecum.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Yuning Zhao
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jie Chen
- Department of Pediatric Cardiology Nephrology and Rheumatism, The Affiliated Hospital of Qingdao University Medical College, Qingdao, 266003, China
| | - Haiting Hu
- Department of Neonatology, The Affiliated Changzhou Maternity and Child Health Care Hospital of Nanjing Medical University, Changzhou, 213004, China
| | - Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214022, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
25
|
Qin D, Ma Y, Wang Y, Hou X, Yu L. Contribution of Lactobacilli on Intestinal Mucosal Barrier and Diseases: Perspectives and Challenges of Lactobacillus casei. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111910. [PMID: 36431045 PMCID: PMC9696601 DOI: 10.3390/life12111910] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The intestine barrier, the front line of normal body defense, relies on its structural integrity, microbial composition and barrier immunity. The intestinal mucosal surface is continuously exposed to a complex and dynamic community of microorganisms. Although it occupies a relatively small proportion of the intestinal microbiota, Lactobacilli has been discovered to have a significant impact on the intestine tract in previous studies. It is undeniable that some Lactobacillus strains present probiotic properties through maintaining the micro-ecological balance via different mechanisms, such as mucosal barrier function and barrier immunity, to prevent infection and even to solve some neurology issues by microbiota-gut-brain/liver/lung axis communication. Notably, not only living cells but also Lactobacillus derivatives (postbiotics: soluble secreted products and para-probiotics: cell structural components) may exert antipathogenic effects and beneficial functions for the gut mucosal barrier. However, substantial research on specific effects, safety and action mechanisms in vivo should be done. In clinical application of humans and animals, there are still doubts about the precise evaluation of Lactobacilli's safety, therapeutic effect, dosage and other aspects. Therefore, we provide an overview of central issues on the impacts of Lactobacillus casei (L. casei) and their products on the intestinal mucosal barrier and some diseases and highlight the urgent need for further studies.
Collapse
Affiliation(s)
- Da Qin
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yixuan Ma
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yanhong Wang
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xilin Hou
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| | - Liyun Yu
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| |
Collapse
|
26
|
Gut microbiota: a new avenue to reveal pathological mechanisms of constipation. Appl Microbiol Biotechnol 2022; 106:6899-6913. [PMID: 36190540 DOI: 10.1007/s00253-022-12197-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
Constipation is very pervasive all over the world. It is a common multifactorial gastrointestinal disease, and its etiology and pathomechanism are not completely clear. Now, increasing evidence shows that intestinal flora is closely related to constipation. Intestinal flora is the largest microbiota in the human body and has powerful metabolic functions. Intestinal flora can produce a variety of metabolites, such as bile acids, short-chain fatty acids, tryptophan metabolites, and methane, which have important effects on intestinal motility and secretion. The host can also monitor the intestinal flora and regulate gut dysbacteriosis in constipation. To explore the relationship between intestinal flora and host, the combination of multiomics technology has become the powerful and effective method. Furthermore, the homeostasis restoration of intestinal flora also provides a new strategy for the treatment of constipation. This review aims to explore the interaction between intestinal flora and host in constipation, which contributes to disclose the pathogenesis of constipation and the development of novel drugs for the treatment of constipation from the perspective of intestinal flora. KEY POINTS: • This review highlights the regulation of gut microbiota on the intestinal motility and secretion of host. • The current review gives an insight into the role of the host on the recognition and regulation of intestinal ecology under constipation. • The article also introduces some novel methods of current gut microbiota research and gut microbiota-based constipation therapies.
Collapse
|
27
|
Miteva D, Peshevska-Sekulovska M, Snegarova V, Batselova H, Alexandrova R, Velikova T. Mucosal COVID-19 vaccines: Risks, benefits and control of the pandemic. World J Virol 2022; 11:221-236. [PMID: 36188733 PMCID: PMC9523321 DOI: 10.5501/wjv.v11.i5.221] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/14/2022] [Accepted: 08/10/2022] [Indexed: 02/05/2023] Open
Abstract
Based on mucosal immunization to promote both mucosal and systemic immune responses, next-generation coronavirus disease 2019 (COVID-19) vaccines would be administered intranasally or orally. The goal of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is to provide adequate immune protection and avoid severe disease and death. Mucosal vaccine candidates for COVID-19 including vector vaccines, recombinant subunit vaccines and live attenuated vaccines are under development. Furthermore, subunit protein vac-cines and virus-vectored vaccines have made substantial progress in preclinical and clinical settings, resulting in SARS-CoV-2 intranasal vaccines based on the previously successfully used nasal vaccines. Additional to their ability to trigger stable, protective immune responses at the sites of pathogenic infection, the development of 'specific' mucosal vaccines targeting coronavirus antigens could be an excellent option for preventing future pandemics. However, their efficacy and safety should be confirmed.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Sofia University “St. Kliment Ohridski,” Faculty of Biology, Sofia 1164, Bulgaria
| | - Monika Peshevska-Sekulovska
- Department of Gastroenterology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Violeta Snegarova
- Clinic of Internal Diseases, Naval Hospital - Varna, Military Medical Academy, Medical Faculty, Medical University, Varna 9000, Bulgaria
| | - Hristiana Batselova
- Department of Epidemiology and Disaster Medicine, Medical University, Plovdiv, University Hospital “St George”, Plovdiv 6000, Bulgaria
| | - Radostina Alexandrova
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia 1000, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia 1407, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
28
|
Gutierrez-Camacho JR, Avila-Carrasco L, Martinez-Vazquez MC, Garza-Veloz I, Zorrilla-Alfaro SM, Gutierrez-Camacho V, Martinez-Fierro ML. Oral Lesions Associated with COVID-19 and the Participation of the Buccal Cavity as a Key Player for Establishment of Immunity against SARS-CoV-2. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11383. [PMID: 36141654 PMCID: PMC9517300 DOI: 10.3390/ijerph191811383] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Some oral lesions have been described in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); the possibility has been raised that the buccal lesions observed in patients with the coronavirus disease 2019 (COVID-19) are due to this virus and the patient's systemic condition. The aim of this review was to integrate the knowledge related to the oral lesions associated with COVID-19 and the participation of the buccal cavity in the establishment of immunity against SARS-CoV-2. METHODS A literature search on the manifestations of buccal lesions from the beginning of the pandemic until October 2021 was carried out by using the PubMed database. A total of 157 scientific articles were selected from the library, which included case reports and reports of lesions appearing in patients with COVID-19. RESULTS Oral lesions included erosions, ulcers, vesicles, pustules, plaques, depapillated tongue, and pigmentations, among others. The oral cavity is a conducive environment for the interaction of SARS-CoV-2 with the mucosal immune system and target cells; direct effects of the virus in this cavity worsen the antiviral inflammatory response of underlying oral disorders, immunodeficiencies, and autoimmunity primarily. CONCLUSIONS The oral cavity is an accessible and privileged environment for the interaction of SARS-CoV-2 with the mucosal immune system and target cells; the direct effects of the virus in this cavity worsen the antiviral inflammatory response of underlying oral disorders, in particular those related to immunodeficiencies and autoimmunity.
Collapse
|
29
|
Davis EC, Castagna VP, Sela DA, Hillard MA, Lindberg S, Mantis NJ, Seppo AE, Järvinen KM. Gut microbiome and breast-feeding: Implications for early immune development. J Allergy Clin Immunol 2022; 150:523-534. [PMID: 36075638 PMCID: PMC9463492 DOI: 10.1016/j.jaci.2022.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | | | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Margaret A Hillard
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Samantha Lindberg
- Department of Biomedical Sciences, University of Albany, Rensselaer, NY
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Albany, NY
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
30
|
Loktionov A. Colon mucus in colorectal neoplasia and beyond. World J Gastroenterol 2022; 28:4475-4492. [PMID: 36157924 PMCID: PMC9476883 DOI: 10.3748/wjg.v28.i32.4475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/23/2022] [Accepted: 08/06/2022] [Indexed: 02/06/2023] Open
Abstract
Little was known about mammalian colon mucus (CM) until the beginning of the 21st century. Since that time considerable progress has been made in basic research addressing CM structure and functions. Human CM is formed by two distinct layers composed of gel-forming glycosylated mucins that are permanently secreted by goblet cells of the colonic epithelium. The inner layer is dense and impenetrable for bacteria, whereas the loose outer layer provides a habitat for abundant commensal microbiota. Mucus barrier integrity is essential for preventing bacterial contact with the mucosal epithelium and maintaining homeostasis in the gut, but it can be impaired by a variety of factors, including CM-damaging switch of commensal bacteria to mucin glycan consumption due to dietary fiber deficiency. It is proven that impairments in CM structure and function can lead to colonic barrier deterioration that opens direct bacterial access to the epithelium. Bacteria-induced damage dysregulates epithelial proliferation and causes mucosal inflammatory responses that may expand to the loosened CM and eventually result in severe disorders, including colitis and neoplastic growth. Recently described formation of bacterial biofilms within the inner CM layer was shown to be associated with both inflammation and cancer. Although obvious gaps in our knowledge of human CM remain, its importance for the pathogenesis of major colorectal diseases, comprising inflammatory bowel disease and colorectal cancer, is already recognized. Continuing progress in CM exploration is likely to result in the development of a range of new useful clinical applications addressing colorectal disease diagnosis, prevention and therapy.
Collapse
|
31
|
Perruzza L, Strati F, Raneri M, Li H, Gargari G, Rezzonico-Jost T, Palatella M, Kwee I, Morone D, Seehusen F, Sonego P, Donati C, Franceschi P, Macpherson AJ, Guglielmetti S, Greiff V, Grassi F. Apyrase-mediated amplification of secretory IgA promotes intestinal homeostasis. Cell Rep 2022; 40:111112. [PMID: 35858559 DOI: 10.1016/j.celrep.2022.111112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/15/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
Secretory immunoglobulin A (SIgA) interaction with commensal bacteria conditions microbiota composition and function. However, mechanisms regulating reciprocal control of microbiota and SIgA are not defined. Bacteria-derived adenosine triphosphate (ATP) limits T follicular helper (Tfh) cells in the Peyer's patches (PPs) via P2X7 receptor (P2X7R) and thereby SIgA generation. Here we show that hydrolysis of extracellular ATP (eATP) by apyrase results in amplification of the SIgA repertoire. The enhanced breadth of SIgA in mice colonized with apyrase-releasing Escherichia coli influences topographical distribution of bacteria and expression of genes involved in metabolic versus immune functions in the intestinal epithelium. SIgA-mediated conditioning of bacteria and enterocyte function is reflected by differences in nutrient absorption in mice colonized with apyrase-expressing bacteria. Apyrase-induced SIgA improves intestinal homeostasis and attenuates barrier impairment and susceptibility to infection by enteric pathogens in antibiotic-induced dysbiosis. Therefore, amplification of SIgA by apyrase can be leveraged to restore intestinal fitness in dysbiotic conditions.
Collapse
Affiliation(s)
- Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Francesco Strati
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Matteo Raneri
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Hai Li
- Maurice Müller Laboratories, Department of Biomedical Research, Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, University of Bern, Bern 3010, Switzerland
| | - Giorgio Gargari
- Division of Food Microbiology and Bioprocesses, Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan 20133, Italy
| | - Tanja Rezzonico-Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Martina Palatella
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Ivo Kwee
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Frauke Seehusen
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich 8057, Switzerland
| | - Paolo Sonego
- Unit of Computational Biology, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige (TN) 38098, Italy
| | - Claudio Donati
- Unit of Computational Biology, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige (TN) 38098, Italy
| | - Pietro Franceschi
- Unit of Computational Biology, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige (TN) 38098, Italy
| | - Andrew J Macpherson
- Maurice Müller Laboratories, Department of Biomedical Research, Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, University of Bern, Bern 3010, Switzerland
| | - Simone Guglielmetti
- Division of Food Microbiology and Bioprocesses, Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan 20133, Italy
| | - Victor Greiff
- Department of Immunology and Oslo University Hospital, University of Oslo, Oslo 0372, Norway
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona 6500, Switzerland.
| |
Collapse
|
32
|
Transcytosis of IgA Attenuates Salmonella Invasion in Human Enteroids and Intestinal Organoids. Infect Immun 2022; 90:e0004122. [PMID: 35579465 DOI: 10.1128/iai.00041-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Secretory IgA (SIgA) is the most abundant antibody type in intestinal secretions where it contributes to safeguarding the epithelium from invasive pathogens like the Gram-negative bacterium, Salmonella enterica serovar Typhimurium (STm). For example, we recently reported that passive oral administration of the recombinant monoclonal SIgA antibody, Sal4, to mice promotes STm agglutination in the intestinal lumen and restricts bacterial invasion of Peyer's patch tissues. In this report, we sought to recapitulate Sal4-mediated protection against STm in human Enteroids and human intestinal organoids (HIOs) as models to decipher the molecular mechanisms by which antibodies function in mucosal immunity in the human gastrointestinal tract. We confirm that Enteroids and HIO-derived monolayers are permissive to STm infection, dependent on HilD, the master transcriptional regulator of the SPI-I type three secretion system (T3SS). Stimulation of M-like cells in both Enteroids and HIOs by the addition of RANKL further enhanced STm invasion. The apical addition of Sal4 mouse IgA, as well as recombinant human Sal4 dimeric IgA (dIgA) and SIgA resulted a dose-dependent reduction in bacterial invasion. Moreover, basolateral application of Sal4 dIgA to Enteroid and HIO monolayers gave rise to SIgA in the apical compartment via a pathway dependent on expression of the polymeric immunoglobulin receptor (pIgR). The resulting Sal4 SIgA was sufficient to reduce STm invasion of Enteroid and HIO epithelial cell monolayers by ~20-fold. Recombinant Sal4 IgG was also transported in the Enteroid and HIOs, but to a lesser degree and via a pathway dependent on the neonatal Fc receptor (FCGRT). The models described lay the foundation for future studies into detailed mechanisms of IgA and IgG protection against STm and other pathogens.
Collapse
|
33
|
Liu X, Wang J, Deng H, Zhong X, Li C, Luo Y, Chen L, Zhang B, Wang D, Huang Y, Zhang J, Guo L. In situ analysis of variations of arsenicals, microbiome and transcriptome profiles along murine intestinal tract. JOURNAL OF HAZARDOUS MATERIALS 2022; 427:127899. [PMID: 34876320 DOI: 10.1016/j.jhazmat.2021.127899] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
In situ-based studies on microbiome-host interactions after arsenic exposure are few. In this study, the variations in arsenics, microbiota, and host genes along murine intestinal tracts were determined after arsenic exposure for two months. There was a gradual increase in the concentration of total As (CtAs) in feces from ileum to colon, whereas CtAs in the corresponding tissues were relatively stable. Differences in arsenic levels between feces and tissues were significantly different. The proportion of arsenite (iAsⅢ) in feces gradually decreased, however, it gradually increased in tissues. After arsenic exposure, the diversity and abundance of microbial community and networks in each segment were significantly dysregulated. Notably, 328, 579 and 90 differently expressed genes were detected in ileum, cecum, and colon, respectively. In addition, microbiome and transcriptome analyses showed a significant correlation between the abundance of Faecalibaculum and expressions of Plb1, Hspa1b, Areg and Duoxa2 genes. This implies that they may be involved in arsenic biotransformation. In vitro experiments using Biofidobactrium and Lactobacillus showed that probiotics have arsenic transformation abilities. Therefore, gut microbiome may modulate arsenic accumulation, excretion and detoxification along the digestive tract. Moreover, the abundance and diversity of gut microbiome may be related to the changes in host health.
Collapse
Affiliation(s)
- Xin Liu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Jiating Wang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Hongyu Deng
- Shenzhen Academy of Metrology and Quality Inspection, Shenzhen 518000, China.
| | - Xiaoting Zhong
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Chengji Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Yu Luo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Linkang Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Bin Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Dongbin Wang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Yixiang Huang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Jingjing Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China; Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease, Guangdong Medical University, Zhanjiang 524001, China.
| | - Lianxian Guo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
34
|
Lee AH, Lin CY, Do S, Oba PM, Belchik SE, Steelman AJ, Schauwecker A, Swanson KS. Dietary supplementation with fiber, "biotics," and spray-dried plasma affects apparent total tract macronutrient digestibility and the fecal characteristics, fecal microbiota, and immune function of adult dogs. J Anim Sci 2022; 100:skac048. [PMID: 35180312 PMCID: PMC8956131 DOI: 10.1093/jas/skac048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
A variety of functional ingredients, including fibers, prebiotics, probiotics, and postbiotics may be added to pet foods to support gastrointestinal and immune health. While many of these ingredients have been tested individually, commercial foods often include blends that also require testing. This study was conducted to evaluate the effects of diets containing blends of fibers, "biotics," and/or spray-dried plasma on apparent total tract digestibility (ATTD), stool quality, fecal microbiota and metabolites, and immune health outcomes of adult dogs. A total of 12 healthy adult intact English pointer dogs (6 M, 6 F; age = 6.4 ± 2.0 yr; BW = 25.8 ± 2.6 kg) were used in a replicated 3 × 3 Latin square design to test diets formulated to: 1) contain a low concentration of fermentative substances (control diet, CT); 2) be enriched with a fiber-prebiotic-probiotic blend (FPPB); and 3) be enriched with a fiber-prebiotic-probiotic blend + immune-modulating ingredients (iFFPB). In each 28-d period, 22 d of diet adaptation was followed by a 5-d fecal collection phase and 1 d for blood sample collection. All data were analyzed using SAS 9.4, with significance being P < 0.05 and trends being P < 0.10. FPPB and iFPPB diets led to shifts in numerous outcome measures. Dry matter (DM), organic matter, fat, fiber, and energy ATTD were lower (P < 0.01), fecal scores were lower (P < 0.01; firmer stools), and fecal DM% was higher (P < 0.0001) in dogs fed FPPB or iFPPB than those fed CT. Serum triglycerides and cholesterol were lower (P < 0.01) in dogs fed FPPB or iFPPB than those fed CT. Fecal protein catabolites (isobutyrate, isovalerate, indole, and ammonia) and butyrate were lower (P < 0.05), while fecal immunoglobulin A (IgA) was higher (P < 0.01) in dogs fed FPPB and iFPPB than those fed CT. Fecal microbiota populations were affected by diet, with alpha-diversity being lower (P < 0.05) in dogs fed iFPPB and the relative abundance of 20 bacterial genera being altered in dogs fed FPPB or iFPPB compared with CT. The circulating helper T cell:cytotoxic T cell ratio was higher (P < 0.05) in dogs fed iFPPB than those fed CT. Circulating B cells were lower (P < 0.05) in dogs fed FPPB than those fed iFPPB, and lower (P < 0.05) in dogs fed iFPPB than those fed CT. Our results demonstrate that feeding a fiber-prebiotic-probiotic blend may provide many benefits to canine health, including improved stool quality, beneficial shifts to fecal microbiota and metabolite profiles, reduced blood lipids, and increased fecal IgA.
Collapse
Affiliation(s)
- Anne H Lee
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ching-Yen Lin
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sungho Do
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Patricia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sara E Belchik
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Amy Schauwecker
- PetSmart Proprietary Brand Product Development, Phoenix, AZ 85080, USA
| | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
35
|
Scheithauer TP, Davids M, Winkelmeijer M, Verdoes X, Aydin Ö, de Brauw M, van de Laar A, Meijnikman AS, Gerdes VE, van Raalte D, Herrema H, Nieuwdorp M. Compensatory intestinal antibody response against pro-inflammatory microbiota after bariatric surgery. Gut Microbes 2022; 14:2031696. [PMID: 35130127 PMCID: PMC8824225 DOI: 10.1080/19490976.2022.2031696] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [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
Obesity and type 2 diabetes (T2D) are growing burdens for individuals and the health-care system. Bariatric surgery is an efficient, but drastic treatment to reduce body weight, normalize glucose values, and reduce low-grade inflammation. The gut microbiome, which is in part controlled by intestinal antibodies, such as IgA, is involved in the development of both conditions. Knowledge of the effect of bariatric surgery on systemic and intestinal antibody response is limited. Here, we determined the fecal antibody and gut microbiome response in 40 T2D and non-diabetic (ND) obese individuals that underwent bariatric surgery (N = 40). Body weight, fasting glucose concentrations and inflammatory parameters decreased after bariatric surgery, whereas pro-inflammatory bacterial species such as lipopolysaccharide (LPS), and flagellin increased in the feces. Simultaneously, concentrations of LPS- and flagellin-specific intestinal IgA levels increased with the majority of pro-inflammatory bacteria coated with IgA after surgery. Finally, serum antibodies decreased in both groups, along with a lower inflammatory tone. We conclude that intestinal rearrangement by bariatric surgery leads to expansion of typical pro-inflammatory bacteria, which may be compensated by an improved antibody response. Although further evidence and mechanistic insights are needed, we postulate that this apparent compensatory antibody response might help to reduce systemic inflammation by neutralizing intestinal immunogenic components and thereby enhance intestinal barrier function after bariatric surgery.
Collapse
Affiliation(s)
- Torsten P.M. Scheithauer
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands,CONTACT Torsten P.M. Scheithauer Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Location AMC, Amsterdam, AZ1105, The Netherlands
| | - Mark Davids
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Maaike Winkelmeijer
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Xanthe Verdoes
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Ömrüm Aydin
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands,Department of Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Maurits de Brauw
- Department of Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | | | - Abraham S. Meijnikman
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands,Department of Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Victor E.A. Gerdes
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Daniël van Raalte
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands,Diabetes Center; Department of Internal Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Hilde Herrema
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands,Department of Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands,Diabetes Center; Department of Internal Medicine, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| |
Collapse
|
36
|
Yan D, Qiang Y, Tian T, Lu D, Wu C. The Effect of Endotoxin on the Intestinal Mucus Layer in Non- and Post-pregnancy Mice. Front Vet Sci 2022; 8:824170. [PMID: 35224078 PMCID: PMC8866870 DOI: 10.3389/fvets.2021.824170] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022] Open
Abstract
The intestine is the most extensive storage organ of bacteria and endotoxins, and the mucosal immune system is the first barrier of the intestine. Mucin-2 (MUC2) is the major component of the mucus layers. In this study, we explored whether MUC2 plays a role in how lipopolysaccharide (LPS) invades the fetus from the gut to the uterus in pregnant mice. The results showed that the LPS levels of the ileum, colon, and uterus were significantly increased, and the content of secretory IgA (sIgA) in the ileum, colon, and uterus tissues was significantly decreased in the LPS(+) group on the 35th day after LPS treatment. On the 16th day of pregnancy, compared with the LPS(-) group, the level of ileum LPS was significantly decreased, and the content of LPS in the fetus was significantly increased in the LPS(+) group. The sIgA content in the fetus was significantly decreased in the uterus and placenta. The expression of MUC2 in the uterus, ileum, and colon was increased significantly in the LPS(+) group, especially in the uterus. It is suggested that endotoxins accumulate in the uterus during non-pregnancy. The high expression of MUC2 in the uterus can prevent LPS from translocating into uterine tissue. After pregnancy, MUC2 still protects uterine tissue, allowing a large amount of LPS to enter the fetal body through blood circulation. Therefore, the level of sIgA significantly decreased, resulting in a decline in fetal innate immune function.
Collapse
Affiliation(s)
- Dujian Yan
- AKS Vocational and Technical College, Aksu, China
| | - Yuyun Qiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Tian Tian
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dezhang Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Chenchen Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
37
|
Alu A, Chen L, Lei H, Wei Y, Tian X, Wei X. Intranasal COVID-19 vaccines: From bench to bed. EBioMedicine 2022; 76:103841. [PMID: 35085851 PMCID: PMC8785603 DOI: 10.1016/j.ebiom.2022.103841] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 02/05/2023] Open
Abstract
Currently licensed COVID-19 vaccines are all designed for intramuscular (IM) immunization. However, vaccination today failed to prevent the virus infection through the upper respiratory tract, which is partially due to the absence of mucosal immunity activation. Despite the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, the next generation of COVID-19 vaccine is in demand and intranasal (IN) vaccination method has been demonstrated to be potent in inducing both mucosal and systemic immune responses. Presently, although not licensed, various IN vaccines against SARS-CoV-2 are under intensive investigation, with 12 candidates reaching clinical trials at different phases. In this review, we give a detailed description about current status of IN COVID-19 vaccines, including virus-vectored vaccines, recombinant subunit vaccines and live attenuated vaccines. The ongoing clinical trials for IN vaccines are highlighted. Additionally, the underlying mechanisms of mucosal immunity and potential mucosal adjuvants and nasal delivery devices are also summarized.
Collapse
Affiliation(s)
- Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Boger-May A, Reed T, LaTorre D, Ruley-Haase K, Hoffman H, English L, Roncagli C, Overstreet AM, Boone D. Altered microbial biogeography in an innate model of colitis. Gut Microbes 2022; 14:2123677. [PMID: 36162004 PMCID: PMC9519015 DOI: 10.1080/19490976.2022.2123677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023] Open
Abstract
Changes in the spatial organization, or biogeography, of colonic microbes have been observed in human inflammatory bowel disease (IBD) and mouse models of IBD. We have developed a mouse model of IBD that occurs spontaneously and consistently in the absence of adaptive immunity. Mice expressing tumor necrosis factor-induced protein 3 (TNFAIP3) in intestinal epithelial cells (villin-TNFAIP3) develop colitis when interbred with Recombination Activating 1-deficient mice (RAG1<sup>-/-</sup>). The colitis in villin-TNFAIP3 × RAG1<sup>-/-</sup> (TRAG) mice is prevented by antibiotics, indicating a role for microbes in this innate colitis. We therefore explored the biogeography of microbes and responses to antibiotics in TRAG colitis. Laser capture microdissection and 16S rRNA sequencing revealed altered microbial populations across the transverse axis of the colon as the inner mucus layer of TRAG, but not RAG1<sup>-/-</sup>, mice was infiltrated by microbes, which included increased abundance of the classes Gammaproteobacteria and Actinobacteria. Along the longitudinal axis differences in the efficacy of antibiotics to prevent colitis were evident. Neomycin was most effective for prevention of inflammation in the cecum, while ampicillin was most effective in the proximal and distal colon. RAG1<sup>-/-</sup>, but not TRAG, mice exhibited a structured pattern of bacterial abundance with decreased Firmicutes and Proteobacteria but increased Bacteroidetes along the proximal to distal axis of the gut. TRAG mice exhibited increased relative abundance of potential pathobionts including <i>Bifidobacterium animalis</i> along the longitudinal axis of the gut whereas others, like <i>Helicobacter hepaticus</i> were increased only in the cecum. Potential beneficial organisms including <i>Roseburia</i> were decreased in the proximal regions of the TRAG colon, while <i>Bifidobacterium pseudolongulum</i> was decreased in the TRAG distal colon. Thus, the innate immune system maintains a structured, spatially organized, gut microbiome along the transverse and longitudinal axis of the gut, and disruption of this biogeography is a feature of innate immune colitis.
Collapse
Affiliation(s)
- Antonia Boger-May
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA
| | - Theodore Reed
- Department of Biology, University of Notre Dame, South Bend, IN, USA
| | - Diana LaTorre
- Department of Biology, University of Notre Dame, South Bend, IN, USA
| | - Katelyn Ruley-Haase
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA
| | - Hunter Hoffman
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA
| | - Lauren English
- Department of Biology, University of Notre Dame, South Bend, IN, USA
| | - Connor Roncagli
- Department of Biology, University of Notre Dame, South Bend, IN, USA
| | - Anne-Marie Overstreet
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA
| | - David Boone
- Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA
- Department of Biology, University of Notre Dame, South Bend, IN, USA
| |
Collapse
|
39
|
Liberti A, Leigh BA, Graham Z, Natarajan O, Dishaw LJ. A Role for Secreted Immune Effectors in Microbial Biofilm Formation Revealed by Simple In Vitro Assays. Methods Mol Biol 2022; 2421:127-140. [PMID: 34870816 DOI: 10.1007/978-1-0716-1944-5_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The formation of biofilms is critical for the successful and stable colonization of mucosal surfaces by microbes, which often build three-dimensional environments by exuding exopolysaccharides and other macromolecules such as proteins, lipids, and even DNA. It is not just bacteria, but fungi such as yeast, that form these adherent interacting communities. Historically, biofilms have been studied in the context of pathogenesis, but only recently it has been recognized that important relationships among members of host-associated microbiomes are maintained within the context of biofilms. Host immune responses impact biofilm formation in various ways; for example, it is likely that formation of stable biofilms by non-pathogens improves barrier defenses by not just filling available niche spaces but also by helping to ward off pathogens directly. Recently, it was found that soluble immune effector molecules such as immunoglobulin A (IgA) in mammals serve essential roles in modulating complex biofilm communities in ways that benefit the host. Additional lines of evidence from other secreted immune effectors, such as the variable region-containing chitin-binding proteins (VCBPs) in protochordates, now suggest that this phenomenon is much more widespread than previously recognized. The activity of these immune molecules also likely serves roles beyond those of simple defense strategies; rather, they may be improving the outcome of symbiotic interactions benefiting the host. Thus, traditional immune assays that are aimed at studying the function of secreted immune effectors, such as agglutination assays, should take into account the possibility that the first observation may not be the last if the microbes under study are not directly killed. Here, we describe a series of simple approaches to characterize biofilm formation when bacteria (or yeast) are cultured in the presence of a secreted immune effector. To model this approach, we use microbes isolated from the gut of Ciona robusta, each grown in the presence or absence of VCBPs. The approaches defined here are amenable to diverse model systems and their microbes.
Collapse
Affiliation(s)
- Assunta Liberti
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida, Saint Petersburg, FL, USA
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Brittany A Leigh
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida, Saint Petersburg, FL, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Zachary Graham
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida, Saint Petersburg, FL, USA
| | - Ojas Natarajan
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida, Saint Petersburg, FL, USA
| | - Larry J Dishaw
- Department of Pediatrics, Morsani College of Medicine, Children's Research Institute, University of South Florida, Saint Petersburg, FL, USA.
| |
Collapse
|
40
|
Silverman MA, Green JL. Insight Into Host-Microbe Interactions Using Microbial Flow Cytometry Coupled to Next-Generation Sequencing. J Pediatric Infect Dis Soc 2021; 10:S106-S111. [PMID: 34951471 PMCID: PMC8703255 DOI: 10.1093/jpids/piab092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Antibody-based assays have been a cornerstone of infectious disease diagnostics for over 100 years [1]. These assays rely on the exquisite sensitivity and specificity of humoral response to almost all infections. While next-generation sequencing (NGS) has tremendous potential to improve diagnostics and uncover host-microbial relationships by directly identifying nucleic acids from infectious microbes, challenges and opportunities for new approaches remain. Here, we review a group of cutting-edge techniques that couple antibody responses with flow cytometry of antibody tagged microbes and NGS. These studies are bringing into focus the dynamic relationship between our immune systems and endogenous microbial communities, which are an important source of pathogens. For simplicity, we use the umbrella term mFLOW-Seq (microbial flow cytometry coupled to NGS) to describe these approaches.
Collapse
Affiliation(s)
- Michael A Silverman
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Corresponding Author: Michael A. Silverman, MD, PhD, Department of Pediatrics, The Children’s Hospital of Philadelphia, 3615 Civic Center Blvd. Abramson Research Center, 12th floor, Room 1202, Philadelphia, PA 19104, USA. E-mail:
| | - Jamal L Green
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Biomedical Graduate Studies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Belo GA, Cordeiro BF, Oliveira ER, Braga MP, da Silva SH, Costa BG, Martins FDS, Jan G, Le Loir Y, Gala-García A, Ferreira E, Azevedo V, do Carmo FLR. SlpB Protein Enhances the Probiotic Potential of L. lactis NCDO 2118 in Colitis Mice Model. Front Pharmacol 2021; 12:755825. [PMID: 34987390 PMCID: PMC8721164 DOI: 10.3389/fphar.2021.755825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/29/2021] [Indexed: 01/30/2023] Open
Abstract
Bacteria used in the production of fermented food products have been investigated for their potential role as modulators of inflammation in gastrointestinal tract disorders such as inflammatory bowel diseases (IBD) that cause irreversible changes in the structure and function of gut tissues. Ulcerative colitis (UC) is the most prevalent IBD in the population of Western countries, and it is marked by symptoms such as weight loss, rectal bleeding, diarrhea, shortening of the colon, and destruction of the epithelial layer. The strain Propionibacterium freudenreichii CIRM-BIA 129 recently revealed promising immunomodulatory properties that greatly rely on surface-layer proteins (Slp), notably SlpB. We, thus, cloned the sequence encoding the SlpB protein into the pXIES-SEC expression and secretion vector, and expressed the propionibacterial protein in the lactic acid bacterium Lactococcus lactis NCDO 2118. The probiotic potential of L. lactis NCDO 2118 harboring pXIES-SEC:slpB (L. lactis-SlpB) was evaluated in a UC-mice model induced by Dextran Sulfate Sodium (DSS). During colitis induction, mice receiving L. lactis-SlpB exhibited reduced severity of colitis, with lower weight loss, lower disease activity index, limited shortening of the colon length, and reduced histopathological score, with significant differences, compared with the DSS group and the group treated with L. lactis NCDO 2118 wild-type strain. Moreover, L. lactis-SlpB administration increased the expression of genes encoding tight junction proteins zo-1, cln-1, cln-5, ocln, and muc-2 in the colon, increased IL-10 and TGF-β, and decreased IL-17, TNF-α, and IL-12 cytokines in the colon. Therefore, this work demonstrates that SlpB recombinant protein is able to increase the probiotic potential of the L. lactis strain to alleviate DSS-induced colitis in mice. This opens perspectives for the development of new approaches to enhance the probiotic potential of strains by the addition of SlpB protein.
Collapse
Affiliation(s)
- Giovanna A. Belo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Bárbara F. Cordeiro
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Emiliano R. Oliveira
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Marina P. Braga
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Sara H. da Silva
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Bruno G. Costa
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Flaviano dos S. Martins
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gwénaël Jan
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Yves Le Loir
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Alfonso Gala-García
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
- School of Dentistry, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Enio Ferreira
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fillipe L. R. do Carmo
- Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
- *Correspondence: Fillipe L. R. do Carmo,
| |
Collapse
|
42
|
Sánchez-Salguero E, Corona-Cervantes K, Guzmán-Aquino HA, de la Borbolla-Cruz MF, Contreras-Vargas V, Piña-Escobedo A, García-Mena J, Santos-Argumedo L. Maternal IgA2 Recognizes Similar Fractions of Colostrum and Fecal Neonatal Microbiota. Front Immunol 2021; 12:712130. [PMID: 34804008 PMCID: PMC8601722 DOI: 10.3389/fimmu.2021.712130] [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: 05/19/2021] [Accepted: 10/19/2021] [Indexed: 11/18/2022] Open
Abstract
Microbiota acquired during labor and through the first days of life contributes to the newborn’s immune maturation and development. Mother provides probiotics and prebiotics factors through colostrum and maternal milk to shape the first neonatal microbiota. Previous works have reported that immunoglobulin A (IgA) secreted in colostrum is coating a fraction of maternal microbiota. Thus, to better characterize this IgA-microbiota association, we used flow cytometry coupled with 16S rRNA gene sequencing (IgA-Seq) in human colostrum and neonatal feces. We identified IgA bound bacteria (IgA+) and characterized their diversity and composition shared in colostrum fractions and neonatal fecal bacteria. We found that IgA2 is mainly associated with Bifidobacterium, Pseudomonas, Lactobacillus, and Paracoccus, among other genera shared in colostrum and neonatal fecal samples. We found that metabolic pathways related to epithelial adhesion and carbohydrate consumption are enriched within the IgA2+ fecal microbiota. The association of IgA2 with specific bacteria could be explained because these antibodies recognize common antigens expressed on the surface of these bacterial genera. Our data suggest a preferential targeting of commensal bacteria by IgA2, revealing a possible function of maternal IgA2 in the shaping of the fecal microbial composition in the neonate during the first days of life.
Collapse
Affiliation(s)
- Erick Sánchez-Salguero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Karina Corona-Cervantes
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Hector Armando Guzmán-Aquino
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - María Fernanda de la Borbolla-Cruz
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Víctor Contreras-Vargas
- Department of Gynecology Regional Hospital "October 1st", Institute for Security and Social Services of State Workers (ISSSTE), México City, Mexico
| | - Alberto Piña-Escobedo
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Jaime García-Mena
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
43
|
Dawod B, Marshall JS, Azad MB. Breastfeeding and the developmental origins of mucosal immunity: how human milk shapes the innate and adaptive mucosal immune systems. Curr Opin Gastroenterol 2021; 37:547-556. [PMID: 34634003 PMCID: PMC11451935 DOI: 10.1097/mog.0000000000000778] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Breastfeeding provides passive immunity while the neonatal immune system matures, and may also protect against chronic immune-mediated conditions long after weaning. This review summarizes current knowledge and new discoveries about human milk and mucosal immunity. RECENT FINDINGS New data suggest that certain microbes in maternal milk may seed and shape the infant gut microbiota, which play a key role in regulating gut barrier integrity and training the developing immune system. Human milk oligosaccharides, best known for their prebiotic functions, have now been shown to directly modulate gene expression in mast and goblet cells in the gastrointestinal tract. Epidemiologic data show a reduced risk of peanut sensitization among infants breastfed by peanut-consuming mothers, suggesting a role for milk-borne food antigens in tolerance development. Cross-fostering experiments in mice suggest the soluble Toll-like receptor 2, found in human milk, may be critical in this process. Finally, interest in human milk antibodies surged during the pandemic with the identification of neutralizing severe acute respiratory syndrome coronavirus 2 antibodies in maternal milk following both natural infection and vaccination. SUMMARY Human milk provides critical immune protection and stimulation to breastfed infants. Understanding the underlying mechanisms could identify new therapeutic targets and strategies for disease prevention across the lifespan.
Collapse
Affiliation(s)
- Bassel Dawod
- Department of Pathology
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia
| | - Jean S. Marshall
- Department of Pathology
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia
| | - Meghan B. Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba
- Department of Pediatrics and Child Health
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
44
|
Brown H, Esterházy D. Intestinal immune compartmentalization: implications of tissue specific determinants in health and disease. Mucosal Immunol 2021; 14:1259-1270. [PMID: 34211125 DOI: 10.1038/s41385-021-00420-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/05/2021] [Accepted: 05/24/2021] [Indexed: 02/04/2023]
Abstract
The emerging concept of tissue specific immunity has opened the gates to new inquiries into what factors drive immune cell niche adaptation and the implications on immune homeostasis, organ specific immune diseases, and therapeutic efficacy. These issues are particularly complicated at barrier sites, which are directly exposed to an ever-changing environment. In particular, the gastrointestinal (GI) tract faces even further challenges given the profound functional and structural differences along its length, raising the possibility that it may even have to be treated as multiple organs when seeking to answer these questions. In this review, we evaluate what is known about the tissue intrinsic and extrinsic factors shaping immune compartments in the intestine. We then discuss the physiological and pathological consequences of a regionally distinct immune system in a single organ, but also discuss where our insight into the role of the compartment for disease development is still very limited. Finally, we discuss the technological and therapeutic implications this compartmentalization has. While the gut is perhaps one of the most intensely studied systems, many of these aspects apply to understanding tissue specific immunity of other organs, most notably other barrier sites such as skin, lung, and the urogenital tract.
Collapse
Affiliation(s)
- Hailey Brown
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Daria Esterházy
- Committee on Immunology, University of Chicago, Chicago, IL, USA. .,Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
45
|
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: 5.7] [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
|
46
|
Chu ND, Crothers JW, Nguyen LTT, Kearney SM, Smith MB, Kassam Z, Collins C, Xavier R, Moses PL, Alm EJ. Dynamic Colonization of Microbes and Their Functions after Fecal Microbiota Transplantation for Inflammatory Bowel Disease. mBio 2021; 12:e0097521. [PMID: 34281401 PMCID: PMC8406238 DOI: 10.1128/mbio.00975-21] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
For fecal microbiota transplantation (FMT) to be successful in immune diseases like inflammatory bowel disease, it is assumed that therapeutic microbes and their beneficial functions and immune interactions must colonize a recipient patient and persist in sufficient quantity and for a sufficient period of time to produce a clinical benefit. Few studies, however, have comprehensively profiled the colonization and persistence of transferred microbes along with the transfer of their microbial functions and interactions with the host immune system. Using 16S, metagenomic, and immunoglobulin A (IgA) sequencing, we analyzed hundreds of longitudinal microbiome samples from a randomized controlled trial of 12 patients with ulcerative colitis who received fecal transplant or placebo for 12 weeks. We uncovered diverse competitive dynamics among donor and patient strains, showing that persistence of transferred microbes is far from static. Indeed, one patient experienced a dramatic loss of donor bacteria 10 weeks into the trial, coinciding with a bloom of pathogenic bacteria and worsening symptoms. We evaluated the transfer of microbial functions, including desired ones, such as butyrate production, and unintended ones, such as antibiotic resistance. By profiling bacteria coated with IgA, we identified bacteria associated with inflammation and found that microbial interactions with the host immune system can be transferred across people, which could play a role in gut microbiome therapeutics for immune-related diseases. Our findings shed light on the colonization dynamics of gut microbes and their functions in the context of FMT to treat a complex disease-information that may provide a foundation for developing more-targeted therapeutics. IMPORTANCE Fecal microbiota transplantation (FMT)-transferring fecal microbes from a healthy donor to a sick patient-has shown promise for gut diseases such as inflammatory bowel disease. Unlike pharmaceuticals, however, fecal transplants are complex mixtures of living organisms, which must then interact with the microbes and immune system of the recipient. We sought to understand these interactions by tracking the microbes of 12 inflammatory bowel disease patients who received fecal transplants for 12 weeks. We uncovered a range of dynamics. For example, one patient experienced successful transfer of donor bacteria, only to lose them after 10 weeks. We similarly evaluated transfer of microbial functions, including how they interacted with the recipient's immune system. Our findings shed light on the colonization dynamics of gut microbes, as well as their functions in the context of FMT-information that may provide a critical foundation for the development of more-targeted therapeutics.
Collapse
Affiliation(s)
- Nathaniel D. Chu
- Center for Microbiome Informatics and Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Le T. T. Nguyen
- Center for Microbiome Informatics and Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Sean M. Kearney
- Center for Microbiome Informatics and Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Zain Kassam
- Finch Therapeutics, Somerville, Massachusetts, USA
| | - Cheryl Collins
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Ramnik Xavier
- Center for Microbiome Informatics and Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Peter L. Moses
- Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Eric J. Alm
- Center for Microbiome Informatics and Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
47
|
Liu Y, Huang W, Wang J, Ma J, Zhang M, Lu X, Liu J, Kou Y. Multifaceted Impacts of Periodontal Pathogens in Disorders of the Intestinal Barrier. Front Immunol 2021; 12:693479. [PMID: 34386004 PMCID: PMC8353228 DOI: 10.3389/fimmu.2021.693479] [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: 04/11/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Periodontal disease, a common inflammatory disease, is considered a hazardous factor that contributes to the development of diseases of the digestive system as well as other systems. The bridge between periodontitis and systemic diseases is believed to be periodontal pathogens. The intestine, as part of the lower gastrointestinal tract, has a close connection with the oral cavity. Within the intestine, the intestinal barrier acts as a multifunctional system including microbial, mucous, physical and immune barrier. The intestinal barrier forms the body's first line of defense against external pathogens; its breakdown can lead to pathological changes in the gut and other organs or systems. Reports in the literature have described how oral periodontal pathogens and pathobiont-reactive immune cells can transmigrate to the intestinal mucosa, causing the destruction of intestinal barrier homeostasis. Such findings might lead to novel ideas for investigating the relationship between periodontal disease and other systemic diseases. This review summarizes studies on the effects of periodontal pathogens on the intestinal barrier, which might contribute to understanding the link between periodontitis and gastrointestinal diseases.
Collapse
Affiliation(s)
- Yingman Liu
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Wenxuan Huang
- School of Stomatology, Shenyang Medical College, Shenyang, China
| | - Jiaqi Wang
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Jiaojiao Ma
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Manman Zhang
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xiaoying Lu
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Jie Liu
- Science Experiment Center, China Medical University, Shenyang, China
| | - Yurong Kou
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
48
|
Optimization of a human milk-directed quantitative sIgA ELISA method substantiated by mass spectrometry. Anal Bioanal Chem 2021; 413:5037-5049. [PMID: 34169348 PMCID: PMC8405464 DOI: 10.1007/s00216-021-03468-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/27/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
Immunoglobulins are the primary protective products in human milk and are responsible for transferring maternal pathogen memory to the infant, providing protection by binding to recognized pathogens and inhibiting virulence. To better understand potentially protective/anti-infective compounds in human milk, the establishment of human milk–tailored analytical approaches is crucial, as most contemporary analytical methods have been optimized for plasma or serum. One of the most prominent immunoglobulins in human milk is secretory immunoglobulin A (sIgA), which may be relevant for the protection of breastfed infants from harmful pathogens. Advanced sIgA detection methods can help monitor the immune status and development of the mother-infant dyad. We therefore developed an enzyme-linked immunosorbent assay (ELISA) sIgA method for the quantitative analysis of IgA plus secretory component (SC), validated with sIgA standards and substantiated by mass spectrometry (MS)–based proteomics. A very strong correlation was observed between the MS-detected IgA1 and the human milk–specific sIgA ELISA (r = 0.82). Overall, the MS data indicate that the developed human milk sIgA ELISA does not differentiate between sIgA1 and sIgA2 and is, therefore, a reflection of total sIgA. Furthermore, our MS data and the human milk–derived sIgA ELISA data are better correlated than data derived from a standard serum IgA ELISA kit (relative to MS IgA1 r = 0.82 and r = 0.42, respectively). We therefore propose our human milk–specific sIgA ELISA as an ideal quantitative indicator of total sIgA with advantages over current serum IgA ELISA kits.
Collapse
|
49
|
Ji Y, Fan X, Zhang Y, Li J, Dai Z, Wu Z. Glycine regulates mucosal immunity and the intestinal microbial composition in weaned piglets. Amino Acids 2021; 54:385-398. [PMID: 33839961 DOI: 10.1007/s00726-021-02976-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
Glycine is an amino acid with a diverse array of health benefits regarding metabolism, immunity, and development. The aim of this study was to test the hypothesis that glycine supplementation alters the intestinal microbial composition and improves the intestinal mucosal immunity of weaned piglets. One hundred and twenty-eight weaned piglets divided into 4 groups were fed with a corn- and soybean meal-based diet supplemented with 0 (control), 0.5, 1, or 2% glycine for 7 days. The intestinal microbiota and tissue samples from the control and the 2% glycine-supplemented piglets were collected for determination of the composition of microbial community and the intestinal mucosal barrier function. Piglets fed with diet containing 2% glycine, instead of 0.5% or 1% glycine, presented elevated average daily gain and feed conversion ratio, as compared with the control. 2% glycine enhanced the abundance of mucins in the jejunum and ileum and mRNA level of porcine β-defensin (pBD) 2 and pBD-3, as well as the protein level of secretory immunoglobulin A (sIgA) in the jejunum. The mRNA expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, and the protein level of phosphorylated p38 mitogen-activated protein kinase (MAPK), signal transducer and activator of transcription 3 (STAT3), nuclear factor (NF)-κB p65, and claudin-2 in the jejunum were lower in the 2% glycine group than that in the control. In addition, an elevated ratio of CD4+/CD8+ T lymphocytes was observed in the jejunum of piglets receiving diet supplemented with 2% glycine. The colon content of piglets fed with 2% glycine exhibited a reduction in abundance of pathogenic bacteria (Escherichia-Shigella, Clostridium, and Burkholderiales) and an increase in short-chain fatty acid-producing bacteria (Blautia, Lachnospiraceae, Anaerostipes, and Prevotella) in comparison with the control. We conclude that dietary supplementation with 2% glycine improves the intestinal immunological barrier function and the microbial composition, therefore, contributing to the growth performance of weaned piglets.
Collapse
Affiliation(s)
- Yun Ji
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Xiaoxiao Fan
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Yunchang Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Ju Li
- Henan Yinfa Animal Husbandry Co., Xinzheng, 451100, Henan, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
50
|
The diverse roles of myeloid derived suppressor cells in mucosal immunity. Cell Immunol 2021; 365:104361. [PMID: 33984533 DOI: 10.1016/j.cellimm.2021.104361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
The mucosal immune system plays a vital role in protecting the host from the external environment. Its major challenge is to balance immune responses against harmful and harmless agents and serve as a 'homeostatic gate keeper'. Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of undifferentiated cells that are characterized by an immunoregulatory and immunosuppressive phenotype. Herein we postulate that MDSCs may be involved in shaping immune responses related to mucosal immunity, due to their immunomodulatory and tissue remodeling functions. Until recently, MDSCs were investigated mainly in cancerous diseases, where they induce and contribute to an immunosuppressive and inflammatory environment that favors tumor development. However, it is now becoming clear that MDSCs participate in non-cancerous conditions such as chronic infections, autoimmune diseases, pregnancy, aging processes and immune tolerance to commensal microbiota at mucosal sites. Since MDSCs are found in the periphery only in small numbers under normal conditions, their role is highlighted during pathologies characterized by acute or chronic inflammation, when they accumulate and become activated. In this review, we describe several aspects of the current knowledge characterizing MDSCs and their involvement in the regulation of the mucosal epithelial barrier, their crosstalk with commensal microbiota and pathogenic microorganisms, and their complex interactions with a variety of surrounding regulatory and effector immune cells. Finally, we discuss the beneficial and harmful outcomes of the MDSC regulatory functions in diseases affecting mucosal tissues. We wish to illuminate the pivotal role of MDSCs in mucosal immunity, the limitations in our understanding of all the players and the intricate challenges stemming from the complex interactions of MDSCs with their environment.
Collapse
|