1
|
Dang S, Zhang X, Zhang Y, Zhang H. New thoughts on the intestinal microbiome-B cell-IgA axis and therapies in IgA nephropathy. Autoimmun Rev 2025:103835. [PMID: 40360014 DOI: 10.1016/j.autrev.2025.103835] [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: 03/21/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
IgA nephropathy (IgAN), as the most common chronic glomerulonephritis worldwide, is often triggered by mucosal infections and follows a chronic progression, with the majority of patients ultimately progressing to end-stage renal disease (ESRD) during their lifetimes. Since the mystery of its complete pathogenesis has not been fully solved, the resulting lack of effective early diagnosis and treatment greatly affects the prognosis of patients. Given the well-defined pathological feature of IgA deposition in the mesangial region, the source and role of pathogenic IgA has been focused on. Starting from the microbiology and immunity of the gut, we systematically review both the physiological and the pathological process of microbiome-B cell-IgA axis, from microbial-induced IgA production to the role of IgA in the intestinal immune milieu, and ultimately end up with the various aspects of microbiome-B cell-IgA axis in the pathogenesis of IgAN as well as the corresponding therapeutic initiatives available. Our retrospective review helps researchers to systematically understand the complex role between intestinal flora dysbiosis and pathogenic IgA in IgAN. This understanding provides a foundation for in-depth explorations to uncover more detailed pathogenic mechanisms and to develop more precise and effective diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Shaoqing Dang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangyu Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yuemiao Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China; Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Lee SK, Kwon JH, Jang JW, Bae SH, Yoon SK, Jung ES, Choi JY. The Critical Role of Regulatory T Cells in Immune Tolerance and Rejection Following Liver Transplantation: Interactions With the Gut Microbiome. Transplantation 2025; 109:784-793. [PMID: 39375899 DOI: 10.1097/tp.0000000000005220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Liver transplantation (LT) is the ultimate treatment for patients with end-stage liver disease or early hepatocellular carcinoma. In the context of LT, because of the unique immunological characteristics of human liver allograft, 5%-20% of selected LT recipients can achieve operational tolerance. Nonetheless, there remains a risk of rejection in LT patients. Maintaining immune homeostasis is thus crucial for improving clinical outcomes in these patients. In mechanism, several immune cells, including dendritic cells, Kupffer cells, myeloid-derived suppressor cells, hepatic stellate cells, regulatory B cells, and CD4 + regulatory T cells (Treg), contribute to achieving tolerance following LT. In terms of Treg, it plays a role in successfully minimizing immunosuppression or achieving tolerance post-LT while also reducing the risk of rejection. Furthermore, the gut microbiome modulates systemic immune functions along the gut-liver axis. Recent studies have explored changes in the microbiome and its metabolites under various conditions, including post-LT, acute rejection, and tolerance. Certain functional microbiomes and metabolites exhibit immunomodulatory functions, such as the augmentation of Treg, influencing immune homeostasis. Therefore, understanding the mechanisms of tolerance in LT, the role of Treg in tolerance and rejection, as well as their interactions with gut microbiome, is vital for the management of LT patients.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- Division of Hepatology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Pathology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
3
|
Jasmine, Baraiya DH, Kavya TT, Mandal A, Chakraborty S, Sathish N, Francis CMR, Binoy Joseph D. Epithelial and mesenchymal compartments of the developing bladder and urethra display spatially distinct gene expression patterns. Dev Biol 2025; 520:155-170. [PMID: 39798644 PMCID: PMC7617630 DOI: 10.1016/j.ydbio.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
The lower urinary tract is comprised of the bladder and urethra and develops from the cloaca, a transient endoderm-derived structure formed from the caudal hindgut. After cloacal septation to form the urogenital sinus and anorectal tract, the bladder gradually develops from the anterior portion of the urogenital sinus while the urethra elongates distally into the genital tubercle. The bladder is a target for regenerative and reconstructive therapies but engineering an impermeable bladder epithelial lining has proven challenging. Urethral epithelial function, including its role as an active immune barrier, is poorly studied and neglected in regenerative therapy. A deeper understanding of epithelial patterning of the urogenital sinus by the surrounding mesenchyme, also accounting for sex-specific differences, can inform regenerative therapies. In this study, we identified spatially distinct genes in the epithelial and mesenchymal compartments of the developing mouse bladder and urethra that could be potential drivers of patterning in the lower urinary tract. Our data revealed spatially restricted domains of transcription factor expression in the epithelium that corresponded with bladder or urethra-specific differentiation. Additionally, we identified the genes Wnt2, Klf4 and Pitx2 that localize to the mesenchyme of the developing bladder and could be potential drivers of bladder differentiation. Our data revealed an increase in the expression of several chemokine genes including Cx3cl1 and Cxcl14 in the developing urethral epithelium that correlated with an increase in epithelial-associated macrophages in the urethra. A survey of sex-specific differences in epithelial and mesenchymal compartments revealed several differentially expressed genes between the male and female urethra but few sex-specific differences in bladder. By comparing spatially distinct gene expression in the developing lower urinary tract, our study provides insights into the divergent differentiation trajectories of the fetal bladder and urethra that establish their adult functions.
Collapse
Affiliation(s)
- Jasmine
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Divyeksha H Baraiya
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - T T Kavya
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Aparna Mandal
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Shreya Chakraborty
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Neha Sathish
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Cynthia Marian Rebecca Francis
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Diya Binoy Joseph
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India.
| |
Collapse
|
4
|
Kulkarni DH, Newberry RD. Antigen Uptake in the Gut: An Underappreciated Piece to the Puzzle? Annu Rev Immunol 2025; 43:571-588. [PMID: 40279313 PMCID: PMC12068241 DOI: 10.1146/annurev-immunol-082523-090154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
The mammalian gut is a vast, diverse, and dynamic single-layer epithelial surface exposed to trillions of microbes, microbial products, and the diet. Underlying this epithelium lies the largest collection of immune cells in the body; these cells encounter luminal substances to generate antigen-specific immune responses characterized by tolerance at homeostasis and inflammation during enteric infections. How the outcomes of antigen-specific tolerance and inflammation are appropriately balanced is a central question in mucosal immunology. Furthermore, how substances large enough to generate antigen-specific responses cross the epithelium and encounter the immune system in homeostasis and during inflammation remains largely unexplored. Here we discuss the challenges presented to the gut immune system, the identified pathways by which luminal substances cross the epithelium, and insights suggesting that the pathways used by substances to cross the epithelium affect the ensuing immune response.
Collapse
Affiliation(s)
- Devesha H Kulkarni
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Rodney D Newberry
- John T. Milliken Department of Medicine, School of Medicine, Washington University, Saint Louis, Missouri, USA;
| |
Collapse
|
5
|
Worley MJ. Salmonella Type III Secretion System Effectors. Int J Mol Sci 2025; 26:2611. [PMID: 40141253 PMCID: PMC11942329 DOI: 10.3390/ijms26062611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Salmonella is estimated to infect between 200 million and over 1 billion people per year. The exact number is not known, as many cases go unreported. Integral to the pathogenesis of Salmonella, as well as numerous other Gram-negative pathogens, is its type III effectors. Salmonella possesses two distinct type III secretion systems, encoded by Salmonella pathogenicity island-1 and Salmonella pathogenicity island-2. Together, they secrete at least 49 type III effectors into host cells that are collectively responsible for many of the virulence attributes of this pathogen. These virulence factors facilitate the invasion of host cells, induce and attenuate inflammation, and change the migratory properties of infected phagocytes, among other things. The effects of all type III effectors on Salmonella virulence are discussed.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
6
|
Hou T, Huang X, Lai J, Zhou D. Intra-tumoral bacteria in breast cancer and intervention strategies. Adv Drug Deliv Rev 2025; 217:115516. [PMID: 39828126 DOI: 10.1016/j.addr.2025.115516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/09/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
The microbiome, consisting of a wide range of both beneficial and harmful microorganisms, is vital to various physiological and pathological processes in the human body, including cancer pathogenesis. Tumor progression is often accompanied by the destruction of the vascular system, allowing bacteria to circulate into the tumor area and flourish in an immunosuppressive environment. Microbes are recognized as significant components of the tumor microenvironment. Recent research has increasingly focused on the role of intra-tumoral bacteria in the onset, progression, and treatment of breast cancer-the most prevalent cancer among women. This review elucidates the potential mechanisms by which intra-tumoral bacteria impact breast cancer and discusses different therapeutic approaches aimed at targeting these bacteria. It provides essential insights for enhancing existing treatment paradigms while paving the way for novel anticancer interventions. As our understanding of the microbiome's intricate relationship with cancer deepens, it opens avenues for groundbreaking strategies that could redefine oncology.
Collapse
Affiliation(s)
- Ting Hou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoling Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiahui Lai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Dongfang Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
7
|
Reuben RC, Torres C. Integrating the milk microbiome signatures in mastitis: milk-omics and functional implications. World J Microbiol Biotechnol 2025; 41:41. [PMID: 39826029 PMCID: PMC11742929 DOI: 10.1007/s11274-024-04242-1] [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: 10/13/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025]
Abstract
Mammalian milk contains a variety of complex bioactive and nutritional components and microorganisms. These microorganisms have diverse compositions and functional roles that impact host health and disease pathophysiology, especially mastitis. The advent and use of high throughput omics technologies, including metagenomics, metatranscriptomics, metaproteomics, metametabolomics, as well as culturomics in milk microbiome studies suggest strong relationships between host phenotype and milk microbiome signatures in mastitis. While single omics studies have undoubtedly contributed to our current understanding of milk microbiome and mastitis, they often provide limited information, targeting only a single biological viewpoint which is insufficient to provide system-wide information necessary for elucidating the biological footprints and molecular mechanisms driving mastitis and milk microbiome dysbiosis. Therefore, integrating a multi-omics approach in milk microbiome research could generate new knowledge, improve the current understanding of the functional and structural signatures of the milk ecosystem, and provide insights for sustainable mastitis control and microbiome management.
Collapse
Affiliation(s)
- Rine Christopher Reuben
- Biology Department, King's College, 133 North River Street, Wilkes-Barre, PA, 18711, USA.
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain.
| | - Carmen Torres
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain
| |
Collapse
|
8
|
Herath HDW, Hu YS. Unveiling nanoparticle-immune interactions: how super-resolution imaging illuminates the invisible. NANOSCALE 2025; 17:1213-1224. [PMID: 39618290 PMCID: PMC12042815 DOI: 10.1039/d4nr03838j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Nanoparticles (NPs) have attracted considerable attention in nanomedicine, particularly in harnessing and manipulating immune cells. However, the current understanding of the interactions between NPs and immune cells at the nanoscale remains limited. Advancing this knowledge guides the design principles of NPs. This review offers a historical perspective on the synergistic evolution of immunology and optical microscopy, examines the current landscape of NP applications in immunology, and explores the advancements in super-resolution imaging techniques, which provide new insights into nanoparticle-immune cell interactions. Key findings from recent studies are discussed, along with challenges and future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Herath D W Herath
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL, 60607-7061, USA.
| | - Ying S Hu
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois Chicago, Chicago, IL, 60607-7061, USA.
| |
Collapse
|
9
|
Ishii Y, Shiota A, Takao T, Yamamoto N, Ogawa T, Jo A, Shinozaki S, Fukuyama S, Koga T, Ito M, Tanaka H, Tamura A, Tsukita S, Matsumoto K, Okamoto I, Kan-O K. Claudin-3 deficiency inhibits allergic responses in an ovalbumin-induced asthma mouse model. Allergol Int 2025:S1323-8930(24)00158-8. [PMID: 39809629 DOI: 10.1016/j.alit.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Affiliation(s)
- Yumiko Ishii
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ayaka Shiota
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Division of Respiratory Medicine, National Hospital Organization Fukuoka National Hospital, Fukuoka, Japan
| | - Tomoaki Takao
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Norio Yamamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Ogawa
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihiro Jo
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Shinozaki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoru Fukuyama
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Respiratory Medicine, National Hospital Organization Omuta National Hospital, Omuta, Japan
| | - Tomoaki Koga
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hiroo Tanaka
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan; Laboratory of Barriology and Cell Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan; Department of Pharmacology, Teikyo University School of Medicine, Tokyo, Japan
| | - Atsushi Tamura
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan; Laboratory of Barriology and Cell Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan; Department of Pharmacology, Teikyo University School of Medicine, Tokyo, Japan
| | - Sachiko Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan; Laboratory of Barriology and Cell Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Koichiro Matsumoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Division of Respirology, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiko Kan-O
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
10
|
Salas-López M, Vélez-Ixta JM, Rojas-Guerrero DL, Piña-Escobedo A, Hernández-Hernández JM, Rangel-Calvillo MN, Pérez-Cruz C, Corona-Cervantes K, Juárez-Castelán CJ, García-Mena J. Human Milk Archaea Associated with Neonatal Gut Colonization and Its Co-Occurrence with Bacteria. Microorganisms 2025; 13:85. [PMID: 39858853 PMCID: PMC11767358 DOI: 10.3390/microorganisms13010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Archaea have been identified as early colonizers of the human intestine, appearing from the first days of life. It is hypothesized that the origin of many of these archaea is through vertical transmission during breastfeeding. In this study, we aimed to characterize the archaeal composition in samples of mother-neonate pairs to observe the potential vertical transmission. We performed a cross-sectional study characterizing the archaeal diversity of 40 human colostrum-neonatal stool samples by next-generation sequencing of V5-V6 16S rDNA libraries. Intra- and inter-sample analyses were carried out to describe the Archaeal diversity in each sample type. Human colostrum and neonatal stools presented similar core microbiota, mainly composed of the methanogens Methanoculleus and Methanosarcina. Beta diversity and metabolic prediction results suggest homogeneity between sample types. Further, the co-occurrence network analysis showed associations between Archaea and Bacteria, which might be relevant for these organisms' presence in the human milk and neonatal stool ecosystems. According to relative abundance proportions, beta diversity, and co-occurrence analyses, the similarities found imply that there is vertical transmission of archaea through breastfeeding. Nonetheless, differential abundances between the sample types suggest other relevant sources for colonizing archaea to the neonatal gut.
Collapse
Affiliation(s)
- Maricarmen Salas-López
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
| | - Juan Manuel Vélez-Ixta
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
| | - Diana Laura Rojas-Guerrero
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Gronostajowa 7, 31-007 Kraków, Poland
| | - Alberto Piña-Escobedo
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
| | - José Manuel Hernández-Hernández
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
| | | | - Claudia Pérez-Cruz
- Departamento de Farmacología, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico;
| | - Karina Corona-Cervantes
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
- Institute for Obesity Research, Monterrey Institute of Technology and Higher Education, Monterrey 64849, Mexico
| | - Carmen Josefina Juárez-Castelán
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
| | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, Mexico City 07360, Mexico; (M.S.-L.); (J.M.V.-I.); or (D.L.R.-G.); (A.P.-E.); (J.M.H.-H.)
| |
Collapse
|
11
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
12
|
Cerovic V, Pabst O, Mowat AM. The renaissance of oral tolerance: merging tradition and new insights. Nat Rev Immunol 2025; 25:42-56. [PMID: 39242920 DOI: 10.1038/s41577-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/09/2024]
Abstract
Oral tolerance is the process by which feeding of soluble proteins induces antigen-specific systemic immune unresponsiveness. Oral tolerance is thought to have a central role in suppressing immune responses to 'harmless' food antigens, and its failure can lead to development of pathologies such as food allergies or coeliac disease. However, on the basis of long-standing experimental observations, the relevance of oral tolerance in human health has achieved new prominence recently following the discovery that oral administration of peanut proteins prevents the development of peanut allergy in at-risk human infants. In this Review, we summarize the new mechanistic insights into three key processes necessary for the induction of tolerance to oral antigens: antigen uptake and transport across the small intestinal epithelial barrier to the underlying immune cells; the processing, transport and presentation of fed antigen by different populations of antigen-presenting cells; and the development of immunosuppressive T cell populations that mediate antigen-specific tolerance. In addition, we consider how related but distinct processes maintain tolerance to bacterial antigens in the large intestine. Finally, we outline the molecular mechanisms and functional consequences of failure of oral tolerance and how these may be modulated to enhance clinical outcomes and prevent disease.
Collapse
Affiliation(s)
- Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany.
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany
| | - Allan McI Mowat
- School of Infection and Immunity, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
13
|
de Carvalho Maquiné L, Dos Santos Almeida Coelho KW, da Silva Gomes MF, Vieira JR, Cavalcante TF, de Souza Carvalho EB, Rufino JPF, de Oliveira AT, de Queiroz Costa Neto P, Pereira JO. In vitro probiotic potential of lactic acid bacteria isolated from the intestines of Muscovy ducks. Braz J Microbiol 2024; 55:4115-4128. [PMID: 39080108 PMCID: PMC11711415 DOI: 10.1007/s42770-024-01474-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/23/2024] [Indexed: 01/11/2025] Open
Abstract
The current study was conducted to isolate, test and characterize molecularly and physiologically lactic acid bacteria from the intestines of Muscovy ducks to evaluate their probiotic potential for poultry farming. Three hundred lactic acid bacteria from the gastrointestinal tract of Muscovy ducks were isolated. The strains were phenotypically characterized by observing cell morphology, performing Gram staining, catalase production, and testing their ability to grow in MRS broth at different temperatures, pH values, NaCl concentrations, bile concentration, and in compatibility tests between strains. Nine strains were selected based on their resilience. Eight strains were identified using molecular techniques. These strains exhibited significant tolerance to acidic pH, bile salts, and NaCl, essential for probiotic function. All isolates inhibited the growth of Salmonella enterica serotype Typhimurium (DT104) and Enteropathogenic Escherichia coli serotype O86:H34 (EPEC), showcasing their antimicrobial potential. Antibiotic susceptibility testing revealed 100% resistance to clindamycin and erythromycin but high susceptibility to ampicillin and vancomycin. Growth was observed at various temperatures, indicating mesophilic characteristics. Compatibility tests confirmed their suitability for probiotic formulations. Genomic analysis identified the strains primarily as Enterococcus. Conclusively, the study identified eight out of nine selected lactic acid bacteria strains from Muscovy ducks as autochthonous probiotics, showing resilience to treatments and compatibility for consortium formulation. These strains are suitable for in vivo testing for potential poultry farming applications. Further research on their molecular mechanisms and in vivo effects is needed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Adriano Teixeira de Oliveira
- Science, and Technology of Amazonas - Center Campus of Manaus, Federal Institute of Education, Manaus City, Amazonas State, Brazil
| | | | - José Odair Pereira
- Faculty of Agrarian Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| |
Collapse
|
14
|
Fröhlich E. Local and systemic effects of microplastic particles through cell damage, release of chemicals and drugs, dysbiosis, and interference with the absorption of nutrients. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:315-344. [PMID: 39324551 DOI: 10.1080/10937404.2024.2406192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Microplastic particles (MPs) have been detected in a variety of environmental samples, including soil, water, food, and air. Cellular studies and animal exposures reported that exposure to MPs composed of different polymers might result in adverse effects at the portal of entry (local) or throughout the body (systemic). The most relevant routes of particle uptake into the body are oral and respiratory exposure. This review describes the various processes that may contribute to the adverse effects of MPs. Only MPs up to 5 µm were found to cross epithelial barriers to a significant extent. However, MPs may also exert a detrimental impact on human health by acting at the epithelial barrier and within the lumen of the orogastrointestinal and respiratory tract. The potential for adverse effects on human health resulting from the leaching, sorption, and desorption of chemicals, as well as the impact of MPs on nutritional status and dysbiosis, are reviewed. In vitro models are suggested as a means of (1) assessing permeation, (2) determining adverse effects on cells of the epithelial barrier, (3) examining influence of digestive fluids on leaching, desorption, and particle properties, and (4) role of microbiota-epithelial cell interactions. The contribution of these mechanisms to human health depends upon exposure levels, which unfortunately have been estimated very differently.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria
| |
Collapse
|
15
|
Spinedi E, Docena GH. Physiopathological Roles of White Adiposity and Gut Functions in Neuroinflammation. Int J Mol Sci 2024; 25:11741. [PMID: 39519291 PMCID: PMC11546880 DOI: 10.3390/ijms252111741] [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: 08/07/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
White adipose tissue (WAT) and the gut are involved in the development of neuroinflammation when an organism detects any kind of injury, thereby triggering metainflammation. In fact, the autonomous nervous system innervates both tissues, although the complex role played by the integrated sympathetic, parasympathetic, and enteric nervous system functions have not been fully elucidated. Our aims were to investigate the participation of inflamed WAT and the gut in neuroinflammation. Firstly, we conducted an analysis into how inflamed peripheral WAT plays a key role in the triggering of metainflammation. Indeed, this included the impact of the development of local insulin resistance and its metabolic consequences, a serious hypothalamic dysfunction that promotes neurodegeneration. Then, we analyzed the gut-brain axis dysfunction involved in neuroinflammation by examining cell interactions, soluble factors, the sensing of microbes, and the role of dysbiosis-related mechanisms (intestinal microbiota and mucosal barriers) affecting brain functions. Finally, we targeted the physiological crosstalk between cells of the brain-WAT-gut axis that restores normal tissue homeostasis after injury. We concluded the following: because any injury can result not only in overall insulin resistance and dysbiosis, which in turn can impact upon the brain, but that a high-risk of the development of neuroinflammation-induced neurodegenerative disorder can also be triggered. Thus, it is imperative to avoid early metainflammation by applying appropriate preventive (e.g., lifestyle and diet) or pharmacological treatments to cope with allostasis and thus promote health homeostasis.
Collapse
Affiliation(s)
- Eduardo Spinedi
- Centro de Endocrinología Experimental y Aplicada (CENEXA-UNLP-CONICET-CICPBA), University of La Plata Medical School, La Plata 1900, Argentina
| | - Guillermo Horacio Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP-UNLP-CONICET-CICPBA), School of Sciences, University of La Plata, La Plata 1900, Argentina
| |
Collapse
|
16
|
Oguro-Igashira E, Murakami M, Mori R, Kuwahara R, Kihara T, Kohara M, Fujiwara M, Motooka D, Okuzaki D, Arase M, Toyota H, Peng S, Ogino T, Kitabatake Y, Morii E, Hirota S, Ikeuchi H, Umemoto E, Kumanogoh A, Takeda K. The pyruvate-GPR31 axis promotes transepithelial dendrite formation in human intestinal dendritic cells. Proc Natl Acad Sci U S A 2024; 121:e2318767121. [PMID: 39432783 PMCID: PMC11536072 DOI: 10.1073/pnas.2318767121] [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: 10/26/2023] [Accepted: 08/30/2024] [Indexed: 10/23/2024] Open
Abstract
The intestinal lumen is rich in gut microbial metabolites that serve as signaling molecules for gut immune cells. G-protein-coupled receptors (GPCRs) sense metabolites and can act as key mediators that translate gut luminal signals into host immune responses. However, the impacts of gut microbe-GPCR interactions on human physiology have not been fully elucidated. Here, we show that GPR31, which is activated by the gut bacterial metabolite pyruvate, is specifically expressed on type 1 conventional dendritic cells (cDC1s) in the lamina propria of the human intestine. Using human induced pluripotent stem cell-derived cDC1s and a monolayer human gut organoid coculture system, we show that cDC1s extend their dendrites toward pyruvate on the luminal side, forming transepithelial dendrites (TED). Accordingly, GPR31 activation via pyruvate enhances the fundamental function of cDC1 by allowing efficient uptake of gut luminal antigens, such as dietary compounds and bacterial particles through TED formation. Our results highlight the role of GPCRs in tuning the human gut immune system according to local metabolic cues.
Collapse
Affiliation(s)
- Eri Oguro-Igashira
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Mari Murakami
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Ryota Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Ryuichi Kuwahara
- Department of Gastroenterological Surgery, Division of Inflammatory Bowel Disease Surgery, Hyogo Medical University, Hyogo663-8501, Japan
| | - Takako Kihara
- Department of Surgical Pathology, Hyogo Medical University, Hyogo663-8501, Japan
| | - Masaharu Kohara
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Makoto Fujiwara
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Daisuke Motooka
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka565-0871, Japan
| | - Daisuke Okuzaki
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka565-0871, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka565-0871, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Osaka565-0871, Japan
| | - Mitsuru Arase
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Hironobu Toyota
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Siyun Peng
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Yasuji Kitabatake
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical University, Hyogo663-8501, Japan
| | - Hiroki Ikeuchi
- Department of Gastroenterological Surgery, Division of Inflammatory Bowel Disease Surgery, Hyogo Medical University, Hyogo663-8501, Japan
| | - Eiji Umemoto
- Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka422-8526, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka565-0871, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka565-0871, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Osaka565-0871, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka565-0871, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka565-0871, Japan
| |
Collapse
|
17
|
Caruso R, Lo BC, Chen GY, Núñez G. Host-pathobiont interactions in Crohn's disease. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-00997-y. [PMID: 39448837 DOI: 10.1038/s41575-024-00997-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
The mammalian intestine is colonized by trillions of microorganisms that are collectively referred to as the gut microbiota. The majority of symbionts have co-evolved with their host in a mutualistic relationship that benefits both. Under certain conditions, such as in Crohn's disease, a subtype of inflammatory bowel disease, some symbionts bloom to cause disease in genetically susceptible hosts. Although the identity and function of disease-causing microorganisms or pathobionts in Crohn's disease remain largely unknown, mounting evidence from animal models suggests that pathobionts triggering Crohn's disease-like colitis inhabit certain niches and penetrate the intestinal tissue to trigger inflammation. In this Review, we discuss the distinct niches occupied by intestinal symbionts and the evidence that pathobionts triggering Crohn's disease live in the mucus layer or near the intestinal epithelium. We also discuss how Crohn's disease-associated mutations in the host disrupt intestinal homeostasis by promoting the penetration and accumulation of pathobionts in the intestinal tissue. Finally, we discuss the potential role of microbiome-based interventions in precision therapeutic strategies for the treatment of Crohn's disease.
Collapse
Affiliation(s)
- Roberta Caruso
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Bernard C Lo
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Grace Y Chen
- Department of Internal Medicine and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Barreto-Duran E, Synowiec A, Szczepański A, Gałuszka-Bulaga A, Węglarczyk K, Baj-Krzyworzeka M, Siedlar M, Bochenek M, Dufva M, Dogan AA, Lenart M, Pyrc K. Development of an intestinal mucosa ex vivo co-culture model to study viral infections. J Virol 2024; 98:e0098724. [PMID: 39212448 PMCID: PMC11495016 DOI: 10.1128/jvi.00987-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
Studying viral infections necessitates well-designed cell culture models to deepen our understanding of diseases and develop effective treatments. In this study, we present a readily available ex vivo 3D co-culture model replicating the human intestinal mucosa. The model combines fully differentiated human intestinal epithelium (HIE) with human monocyte-derived macrophages (hMDMs) and faithfully mirrors the in vivo structural and organizational properties of intestinal mucosal tissues. Specifically, it mimics the lamina propria, basement membrane, and the air-exposed epithelial layer, enabling the pioneering observation of macrophage migration through the tissue to the site of viral infection. In this study, we applied the HIE-hMDMs model for the first time in viral infection studies, infecting the model with two globally significant viruses: severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human norovirus GII.4. The results demonstrate the model's capability to support the replication of both viruses and show the antiviral role of macrophages, determined by their migration to the infection site and subsequent direct contact with infected epithelial cells. In addition, we evaluated the production of cytokines and chemokines in the intestinal niche, observing an increased interleukin-8 production during infection. A parallel comparison using a classical in vitro cell line model comprising Caco-2 and THP-1 cells for SARS-CoV-2 experiments confirmed the utility of the HIE-hMDMs model in viral infection studies. Our data show that the ex vivo tissue models hold important implications for advances in virology research.IMPORTANCEThe fabrication of intricate ex vivo tissue models holds important implications for advances in virology research. The co-culture model presented here provides distinct spatial and functional attributes not found in simplified models, enabling the evaluation of macrophage dynamics under severe acute respiratory syndrome coronavirus 2 and human norovirus (HuNoV) infections in the intestine. Moreover, these models, comprised solely of primary cells, facilitate the study of difficult-to-replicate viruses such as HuNoV, which cannot be studied in cell line models, and offer the opportunity for personalized treatment evaluations using patient cells. Similar co-cultures have been established for the study of bacterial infections and different characteristics of the intestinal tissue. However, to the best of our knowledge, a similar intestinal model for the study of viral infections has not been published before.
Collapse
Affiliation(s)
- Emilia Barreto-Duran
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Aleksandra Synowiec
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Artur Szczepański
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Michał Bochenek
- Flow Cytometry Facility, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Martin Dufva
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Asli Aybike Dogan
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marzena Lenart
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
19
|
Kolahi Sadeghi L, Vahidian F, Eterafi M, Safarzadeh E. Gastrointestinal cancer resistance to treatment: the role of microbiota. Infect Agent Cancer 2024; 19:50. [PMID: 39369252 PMCID: PMC11453072 DOI: 10.1186/s13027-024-00605-3] [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: 06/15/2024] [Accepted: 08/20/2024] [Indexed: 10/07/2024] Open
Abstract
The most common illnesses that adversely influence human health globally are gastrointestinal malignancies. The prevalence of gastrointestinal cancers (GICs) is relatively high, and the majority of patients receive ineffective care since they are discovered at an advanced stage of the disease. A major component of the human body is thought to be the microbiota of the gastrointestinal tract and the genes that make up the microbiome. The gut microbiota includes more than 3000 diverse species and billions of microbes. Each of them has benefits and drawbacks and been demonstrated to alter anticancer medication efficacy. Treatment of GIC with the help of the gut bacteria is effective while changes in the gut microbiome which is linked to resistance immunotherapy or chemotherapy. Despite significant studies and findings in this field, more research on the interactions between microbiota and response to treatment in GIC are needed to help researchers provide more effective therapeutic strategies with fewer treatment complication. In this review, we examine the effect of the human microbiota on anti-cancer management, including chemotherapy, immunotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Leila Kolahi Sadeghi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Fatemeh Vahidian
- Faculty of Medicine, Laval University, Quebec, Canada
- Centre de Recherche de I'Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec, Canada
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students' Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology, and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
20
|
Iketani A, Takano M, Kasakura K, Iwatsuki M, Tsuji A, Matsuda K, Minegishi R, Hosono A, Nakanishi Y, Takahashi K. CCAAT/enhancer-binding protein α-dependent regulation of granule formation in mast cells by intestinal bacteria. Eur J Immunol 2024; 54:e2451094. [PMID: 38980255 DOI: 10.1002/eji.202451094] [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: 02/22/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024]
Abstract
The antiallergic effects of gut microbiota have been attracting attention in recent years, but the underlying cellular and molecular mechanisms have not yet been fully understood. In this study, we aimed to investigate these mechanisms specifically focusing on mast cells. Mast cells retain intracellular granules containing various inflammatory mediators such as histamine, which are released outside the cells upon IgE and allergen stimulation. We previously reported that increased expression of the transcription factor, CCAAT/enhancer-binding protein α (C/EBPα), suppresses granule formation in mast cells and that Lacticaseibacillus casei JCM1134T (LC) upregulates C/EBPα levels. Here, granule formation in mouse bone marrow-derived mast cells was suppressed in a MyD88-dependent manner after LC treatment due to C/EBPα-dependent downregulation of the genes encoding serglycin (SRGN) and mast cell protease 4 (Mcpt4). Furthermore, C/EBPα expression was regulated by DNA methylation in the 5' region far upstream of the transcription start site. LC suppressed DNA methylation of specific CpG motifs in the 5' region of the C/EBPα gene. These results conclude that specific gut microbial components, such as those from LC, suppress granule formation in mast cells by inhibiting SRGN and Mcpt4 expression via reduced C/EBPα gene methylation.
Collapse
Affiliation(s)
- Ayaka Iketani
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
| | - Mai Takano
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
| | - Kazumi Kasakura
- Department of Bioresource Utilization Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
| | - Miono Iwatsuki
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Ayu Tsuji
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Kou Matsuda
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Remina Minegishi
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Akira Hosono
- Department of Bioresource Utilization Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Yusuke Nakanishi
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| | - Kyoko Takahashi
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
- Department of Bioresource Utilization Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, Japan
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa, Japan
| |
Collapse
|
21
|
Camarini R, Marianno P, Hanampa-Maquera M, Oliveira SDS, Câmara NOS. Prenatal Stress and Ethanol Exposure: Microbiota-Induced Immune Dysregulation and Psychiatric Risks. Int J Mol Sci 2024; 25:9776. [PMID: 39337263 PMCID: PMC11431796 DOI: 10.3390/ijms25189776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Changes in maternal gut microbiota due to stress and/or ethanol exposure can have lasting effects on offspring's health, particularly regarding immunity, inflammation response, and susceptibility to psychiatric disorders. The literature search for this review was conducted using PubMed and Scopus, employing keywords and phrases related to maternal stress, ethanol exposure, gut microbiota, microbiome, gut-brain axis, diet, dysbiosis, progesterone, placenta, prenatal development, immunity, inflammation, and depression to identify relevant studies in both preclinical and human research. Only a limited number of reviews were included to support the arguments. The search encompassed studies from the 1990s to the present. This review begins by exploring the role of microbiota in modulating host health and disease. It then examines how disturbances in maternal microbiota can affect the offspring's immune system. The analysis continues by investigating the interplay between stress and dysbiosis, focusing on how prenatal maternal stress influences both maternal and offspring microbiota and its implications for susceptibility to depression. The review also considers the impact of ethanol consumption on gut dysbiosis, with an emphasis on the effects of prenatal ethanol exposure on both maternal and offspring microbiota. Finally, it is suggested that maternal gut microbiota dysbiosis may be significantly exacerbated by the combined effects of stress and ethanol exposure, leading to immune system dysfunction and chronic inflammation, which could increase the risk of depression in the offspring. These interactions underscore the potential for novel mental health interventions that address the gut-brain axis, especially in relation to maternal and offspring health.
Collapse
Affiliation(s)
- Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Priscila Marianno
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Maylin Hanampa-Maquera
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Samuel Dos Santos Oliveira
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
22
|
Jensen O, Trujillo E, Hanson L, Ost KS. Controlling Candida: immune regulation of commensal fungi in the gut. Infect Immun 2024; 92:e0051623. [PMID: 38647290 PMCID: PMC11385159 DOI: 10.1128/iai.00516-23] [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] [Indexed: 04/25/2024] Open
Abstract
The intestinal microbiome harbors fungi that pose a significant risk to human health as opportunistic pathogens and drivers of inflammation. Inflammatory and autoimmune diseases are associated with dysbiotic fungal communities and the expansion of potentially pathogenic fungi. The gut is also the main reservoir for disseminated fungal infections. Immune interactions are critical for preventing commensal fungi from becoming pathogenic. Significant strides have been made in defining innate and adaptive immune pathways that regulate intestinal fungi, and these discoveries have coincided with advancements in our understanding of the fungal molecular pathways and effectors involved in both commensal colonization and pathogenesis within the gut. In this review, we will discuss immune interactions important for regulating commensal fungi, with a focus on how specific cell types and effectors interact with fungi to limit their colonization or pathogenic potential. This will include how innate and adaptive immune pathways target fungi and orchestrate antifungal immune responses, in addition to how secreted immune effectors, such as mucus and antimicrobial peptides, regulate fungal colonization and inhibit pathogenic potential. These immune interactions will be framed around our current understanding of the fungal effectors and pathways regulating colonization and pathogenesis within this niche. Finally, we highlight important unexplored mechanisms by which the immune system regulates commensal fungi in the gut.
Collapse
Affiliation(s)
- Owen Jensen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emma Trujillo
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke Hanson
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kyla S. Ost
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
23
|
Gleeson PJ, Monteiro RC. The Role of Mucosal Immunity: What Can We Learn From Animal and Human Studies? Semin Nephrol 2024; 44:151566. [PMID: 40082160 DOI: 10.1016/j.semnephrol.2025.151566] [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: 03/16/2025]
Abstract
Immunoglobulin A (IgA) is a key actor in the mucosal immune system, which moderates interactions between the host and environmental factors such as food antigens and commensal microorganisms. The pathogenesis of IgA nephropathy (IgAN) involves a multistep process starting with deglycosylation of mucosally derived, polymeric IgA1 (dg-IgA1) that reaches the circulation. Modified O-glycans on dg-IgA1 are targeted by IgG-autoantibodies, leading to the formation of circulating immune complexes that deposit in the glomerular mesangium. Infections of mucosal surfaces trigger flares of primary IgAN, while inflammatory bowel disease and liver cirrhosis are important causes of secondary IgAN, supporting a mucosal source of nephritogenic IgA1. In the presence of microbial pathogens or food antigens, activated dendritic cells in the gut mucosa induce T-cell-dependent or T-cell-independent B-cell differentiation into IgA-secreting plasma cells. Herein we review the literature concerning mucosal immune function and how it is altered in this disease. We discuss recent evidence supporting a causal role of gut microbiota dysbiosis in IgAN pathogenesis.
Collapse
Affiliation(s)
- Patrick J Gleeson
- Paris Cité University, Center for Research on Inflammation, Paris, France; Inserm, UMR1149; CNRS EMR8252; Inflamex Laboratory of Excellence; Nephrology Department.
| | - Renato C Monteiro
- Paris Cité University, Center for Research on Inflammation, Paris, France; Inserm, UMR1149; CNRS EMR8252; Inflamex Laboratory of Excellence; Immunology laboratory of Bichat hospital, Paris, France
| |
Collapse
|
24
|
Link K, Muhandes L, Polikarpova A, Lämmermann T, Sixt M, Fässler R, Roers A. Integrin β1-mediated mast cell immune-surveillance of blood vessel content. J Allergy Clin Immunol 2024; 154:745-753. [PMID: 38636606 DOI: 10.1016/j.jaci.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND IgE-mediated degranulation of mast cells (MCs) provides rapid protection against environmental hazards, including animal venoms. A fraction of tissue-resident MCs intimately associates with blood vessels. These perivascular MCs were reported to extend projections into the vessel lumen and to be the first MCs to acquire intravenously injected IgE, suggesting that IgE loading of MCs depends on their vascular association. OBJECTIVE We sought to elucidate the molecular basis of the MC-blood vessel interaction and to determine its relevance for IgE-mediated immune responses. METHODS We selectively inactivated the Itgb1 gene, encoding the β1 chain of integrin adhesion molecules (ITGB1), in MCs by conditional gene targeting in mice. We analyzed skin MCs for blood vessel association, surface IgE density, and capability to bind circulating antibody specific for MC surface molecules, as well as in vivo responses to antigen administered via different routes. RESULTS Lack of ITGB1 expression severely compromised MC-blood vessel association. ITGB1-deficient MCs showed normal densities of surface IgE but reduced binding of intravenously injected antibodies. While their capacity to degranulate in response to IgE ligation in vivo was unimpaired, anaphylactic responses to antigen circulating in the vasculature were largely abolished. CONCLUSIONS ITGB1-mediated association of MCs with blood vessels is key for MC immune surveillance of blood vessel content, but is dispensable for slow steady-state loading of endogenous IgE onto tissue-resident MCs.
Collapse
Affiliation(s)
- Kristina Link
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Lina Muhandes
- Institute for Clinical Chemistry and Clinical Pharmacology, Universitätsklinikum Bonn, Bonn, Germany
| | | | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Michael Sixt
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | | | - Axel Roers
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
25
|
Furuta S. Microbiome-Stealth Regulator of Breast Homeostasis and Cancer Metastasis. Cancers (Basel) 2024; 16:3040. [PMID: 39272898 PMCID: PMC11394247 DOI: 10.3390/cancers16173040] [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: 08/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cumulative evidence attests to the essential roles of commensal microbes in the physiology of hosts. Although the microbiome has been a major research subject since the time of Luis Pasteur and William Russell over 140 years ago, recent findings that certain intracellular bacteria contribute to the pathophysiology of healthy vs. diseased tissues have brought the field of the microbiome to a new era of investigation. Particularly, in the field of breast cancer research, breast-tumor-resident bacteria are now deemed to be essential players in tumor initiation and progression. This is a resurrection of Russel's bacterial cause of cancer theory, which was in fact abandoned over 100 years ago. This review will introduce some of the recent findings that exemplify the roles of breast-tumor-resident microbes in breast carcinogenesis and metastasis and provide mechanistic explanations for these phenomena. Such information would be able to justify the utility of breast-tumor-resident microbes as biomarkers for disease progression and therapeutic targets.
Collapse
Affiliation(s)
- Saori Furuta
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA;
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
26
|
Brychka D, Ayala-Nunez NV, Dupas A, Bare Y, Partiot E, Mittelheisser V, Lucansky V, Goetz JG, Osmani N, Gaudin R. Targeting monocytic Occludin impairs transendothelial migration and HIV neuroinvasion. EMBO Rep 2024; 25:3276-3299. [PMID: 39039298 PMCID: PMC11315906 DOI: 10.1038/s44319-024-00190-x] [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: 10/12/2023] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Transmigration of circulating monocytes from the bloodstream to tissues represents an early hallmark of inflammation. This process plays a pivotal role during viral neuroinvasion, encephalitis, and HIV-associated neurocognitive disorders. How monocytes locally unzip endothelial tight junction-associated proteins (TJAPs), without perturbing impermeability, to reach the central nervous system remains poorly understood. Here, we show that human circulating monocytes express the TJAP Occludin (OCLN) to promote transmigration through endothelial cells. We found that human monocytic OCLN (hmOCLN) clusters at monocyte-endothelium interface, while modulation of hmOCLN expression significantly impacts monocyte transmigration. Furthermore, we designed OCLN-derived peptides targeting its extracellular loops (EL) and show that transmigration of treated monocytes is inhibited in vitro and in zebrafish embryos, while preserving vascular integrity. Monocyte transmigration toward the brain is an important process for HIV neuroinvasion and we found that the OCLN-derived peptides significantly inhibit HIV dissemination to cerebral organoids. In conclusion, our study identifies an important role for monocytic OCLN during transmigration and provides a proof-of-concept for the development of mitigation strategies to prevent monocyte infiltration and viral neuroinvasion.
Collapse
Affiliation(s)
- Diana Brychka
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Montpellier, France
- Univ Montpellier, Montpellier, France
| | - Nilda Vanesa Ayala-Nunez
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Montpellier, France
- Univ Montpellier, Montpellier, France
- Empa - Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Amandine Dupas
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Yonis Bare
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Montpellier, France
- Univ Montpellier, Montpellier, France
| | - Emma Partiot
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Montpellier, France
- Univ Montpellier, Montpellier, France
| | - Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Vincent Lucansky
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Montpellier, France
- Univ Montpellier, Montpellier, France
- Jessenius Faculty of Medicine in Martin (JFMED CU), Department of Pathophysiology, Comenius University in Bratislava, Martin, Slovakia
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Naël Osmani
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Raphael Gaudin
- CNRS, Institut de Recherche en Infectiologie de Montpellier (IRIM), Montpellier, France.
- Univ Montpellier, Montpellier, France.
| |
Collapse
|
27
|
Hummel G, Aagaard K. Arthropods to Eutherians: A Historical and Contemporary Comparison of Sparse Prenatal Microbial Communities Among Animalia Species. Am J Reprod Immunol 2024; 92:e13897. [PMID: 39140417 DOI: 10.1111/aji.13897] [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: 04/24/2023] [Revised: 04/08/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Since the advent of next-generation sequencing, investigators worldwide have sought to discern whether a functional and biologically or clinically relevant prenatal microbiome exists. One line of research has led to the hypothesis that microbial DNA detected in utero/in ovo or prior to birth/hatching is a result of contamination and does not belong to viable and functional microbes. Many of these preliminary evaluations have been conducted in humans, mice, and nonhuman primates due to sample and specimen availability. However, a comprehensive review of the literature across animal species suggests organisms that maintain an obligate relationship with microbes may act as better models for interrogating the selective pressures placed on vertical microbial transfer over traditional laboratory species. To date, studies in humans and viviparous laboratory species have failed to illustrate the clear presence and transfer of functional microbes in utero. Until a ground truth regarding the status and relevance of prenatal microbes can be ascertained, it is salient to conduct parallel investigations into the prevalence of a functional prenatal microbiome across the developmental lifespan of multiple organisms in the kingdom Animalia. This comprehensive understanding is necessary not only to determine the role of vertically transmitted microbes and their products in early human health but also to understand their full One Health impact. This review is among the first to compile such comprehensive primary conclusions from the original investigator's conclusions, and hence collectively illustrates that prenatal microbial transfer is supported by experimental evidence arising from over a long and rigorous scientific history encompassing a breadth of species from kingdom Animalia.
Collapse
Affiliation(s)
- Gwendolynn Hummel
- Departments of Obstetrics and Gynecology (Division of Maternal-Fetal Medicine) and Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Kjersti Aagaard
- Departments of Obstetrics and Gynecology (Division of Maternal-Fetal Medicine) and Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
28
|
Maranha A, Alarico S, Nunes-Costa D, Melo-Marques I, Roxo I, Castanheira P, Caramelo O, Empadinhas N. Drinking Water Microbiota, Entero-Mammary Pathways, and Breast Cancer: Focus on Nontuberculous Mycobacteria. Microorganisms 2024; 12:1425. [PMID: 39065193 PMCID: PMC11279143 DOI: 10.3390/microorganisms12071425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The prospect of drinking water serving as a conduit for gut bacteria, artificially selected by disinfection strategies and a lack of monitoring at the point of use, is concerning. Certain opportunistic pathogens, notably some nontuberculous mycobacteria (NTM), often exceed coliform bacteria levels in drinking water, posing safety risks. NTM and other microbiota resist chlorination and thrive in plumbing systems. When inhaled, opportunistic NTM can infect the lungs of immunocompromised or chronically ill patients and the elderly, primarily postmenopausal women. When ingested with drinking water, NTM often survive stomach acidity, reach the intestines, and migrate to other organs using immune cells as vehicles, potentially colonizing tumor tissue, including in breast cancer. The link between the microbiome and cancer is not new, yet the recognition of intratumoral microbiomes is a recent development. Breast cancer risk rises with age, and NTM infections have emerged as a concern among breast cancer patients. In addition to studies hinting at a potential association between chronic NTM infections and lung cancer, NTM have also been detected in breast tumors at levels higher than normal adjacent tissue. Evaluating the risks of continued ingestion of contaminated drinking water is paramount, especially given the ability of various bacteria to migrate from the gut to breast tissue via entero-mammary pathways. This underscores a pressing need to revise water safety monitoring guidelines and delve into hormonal factors, including addressing the disproportionate impact of NTM infections and breast cancer on women and examining the potential health risks posed by the cryptic and unchecked microbiota from drinking water.
Collapse
Affiliation(s)
- Ana Maranha
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Susana Alarico
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniela Nunes-Costa
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Inês Melo-Marques
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Inês Roxo
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Olga Caramelo
- Gynecology Department, Coimbra Hospital and University Centre (CHUC), 3004-561 Coimbra, Portugal;
| | - Nuno Empadinhas
- Center for Neuroscience and Cell Biology (CNC-UC), University of Coimbra, 3004-504 Coimbra, Portugal; (A.M.); (S.A.); (D.N.-C.); (I.M.-M.); (I.R.)
- Centre for Innovative Biomedicine & Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
29
|
Zhou Y, Zhang D, Cheng H, Wu J, Liu J, Feng W, Peng C. Repairing gut barrier by traditional Chinese medicine: roles of gut microbiota. Front Cell Infect Microbiol 2024; 14:1389925. [PMID: 39027133 PMCID: PMC11254640 DOI: 10.3389/fcimb.2024.1389925] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Gut barrier is not only part of the digestive organ but also an important immunological organ for the hosts. The disruption of gut barrier can lead to various diseases such as obesity and colitis. In recent years, traditional Chinese medicine (TCM) has gained much attention for its rich clinical experiences enriched in thousands of years. After orally taken, TCM can interplay with gut microbiota. On one hand, TCM can modulate the composition and function of gut microbiota. On the other hand, gut microbiota can transform TCM compounds. The gut microbiota metabolites produced during the actions of these interplays exert noticeable pharmacological effects on the host especially gut barrier. Recently, a large number of studies have investigated the repairing and fortifying effects of TCM on gut barriers from the perspective of gut microbiota and its metabolites. However, no review has summarized the mechanism behand this beneficiary effects of TCM. In this review, we first briefly introduce the unique structure and specific function of gut barrier. Then, we summarize the interactions and relationship amidst gut microbiota, gut microbiota metabolites and TCM. Further, we summarize the regulative effects and mechanisms of TCM on gut barrier including physical barrier, chemical barrier, immunological barrier, and microbial barrier. At last, we discuss the effects of TCM on diseases that are associated gut barrier destruction such as ulcerative colitis and type 2 diabetes. Our review can provide insights into TCM, gut barrier and gut microbiota.
Collapse
Affiliation(s)
- Yaochuan Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dandan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlu Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
30
|
Alemu BK, Lee MW, Leung MBW, Lee WF, Wang Y, Wang CC, Lau SL. Preventive effect of prenatal maternal oral probiotic supplementation on neonatal jaundice (POPS Study): A protocol for the randomised double-blind placebo-controlled clinical trial. BMJ Open 2024; 14:e083641. [PMID: 38851232 PMCID: PMC11163667 DOI: 10.1136/bmjopen-2023-083641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 05/21/2024] [Indexed: 06/10/2024] Open
Abstract
INTRODUCTION Neonatal jaundice is a common and life-threatening health problem in neonates due to overaccumulation of circulating unconjugated bilirubin. Gut flora has a potential influence on bilirubin metabolism. The infant gut microbiome is commonly copied from the maternal gut. During pregnancy, due to changes in dietary habits, hormones and body weight, maternal gut dysbiosis is common, which can be stabilised by probiotics supplementation. However, whether probiotic supplements can reach the baby through the mother and reduce the incidence of neonatal jaundice has not been studied yet. Therefore, we aim to evaluate the effect of prenatal maternal probiotic supplementation on the incidence of neonatal jaundice. METHODS AND ANALYSIS This is a randomised double-blind placebo-controlled clinical trial among 94 pregnant women (47 in each group) in a tertiary hospital in Hong Kong. Voluntary eligible participants will be recruited between 28 and 35 weeks of gestation. Computer-generated randomisation and allocation to either the intervention or control group will be carried out. Participants will take either one sachet of Vivomixx (450 billion colony-forming units per sachet) or a placebo per day until 1 week post partum. Neither the study participants nor researchers will know the randomisation and allocation. The intervention will be initiated at 36 weeks of gestation. Neonatal bilirubin level will be measured to determine the primary outcome (hyperbilirubinaemia) while the metagenomic microbiome profile of breast milk and maternal and infant stool samples as well as pregnancy outcomes will be secondary outcomes. Binary logistic and linear regressions will be carried out to assess the association of the microbiome data with different clinical outcomes. ETHICS AND DISSEMINATION Ethics approval is obtained from the Joint CUHK-NTEC Clinical Research Ethics Committee, Hong Kong (CREC Ref: 2023.100-T). Findings will be published in peer-reviewed journals and presented at international conferences. TRIAL REGISTRATION NUMBER NCT06087874.
Collapse
Affiliation(s)
- Bekalu Kassie Alemu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
- Department of Midwifery, College of Medicine and Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - May Wing Lee
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Maran Bo Wah Leung
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Wing Fong Lee
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Yao Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
- Institute of Health Sciences, The Chinese University, Hong Kong, Hong Kong SAR
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
- School of Biomedical Sciences, Joint Laboratory for Reproductive Medicine, The Chinese University, Hong Kong, Hong Kong SAR
| | - So Ling Lau
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| |
Collapse
|
31
|
Wang Y, Zhao Y, Tang X, Nan X, Jiang L, Wang H, Liu J, Yang L, Yao J, Xiong B. Nutrition, gastrointestinal microorganisms and metabolites in mastitis occurrence and control. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:220-231. [PMID: 38800734 PMCID: PMC11126769 DOI: 10.1016/j.aninu.2024.01.010] [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: 03/11/2023] [Revised: 01/02/2024] [Accepted: 01/23/2024] [Indexed: 05/29/2024]
Abstract
Mastitis affects almost all mammals including humans and dairy cows. In the dairy industry, bovine mastitis is a disease with a persistently high incidence, causing serious losses to the health of cows, the quality of dairy products, and the economy of dairy farms. Although local udder infection caused by the invasion of exogenous pathogens into the mammary gland was considered the main cause of mastitis, evidence has been established and continues to grow, showing that nutrition factors and gastrointestinal microbiome (GM) as well as their metabolites are also involved in the development of mammary inflammatory response. Suboptimal nutrition is recognized as a risk factor for increased susceptibility to mastitis in cattle, in particular the negative energy balance. The majority of data regarding nutrition and bovine mastitis involves micronutrients. In addition, the dysbiotic GM can directly trigger or aggravate mastitis through entero-mammary gland pathway. The decreased beneficial commensal bacteria, lowered bacterial diversity, and increased pathogens as well as proinflammatory metabolites are found in both the milk and gastrointestinal tract of mastitic dairy cows. This review discussed the relationship between the nutrition (energy and micronutrient levels) and mastitis, summarized the role of GM and metabolites in regulating mastitis. Meanwhile, several non-antibiotics strategies were provided for the prevention and alleviation of mastitis, including micronutrients, probiotics, short-chain fatty acids, high-fiber diet, inulin, and aryl hydrocarbon receptor.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Yiguang Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Linshu Jiang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing 102206, China
| | - Hui Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jun Liu
- Langfang Academy of Agriculture and Forestry, Langfang 065000, China
| | - Liang Yang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
32
|
Han R, He H, Lu Y, Lu H, Shen S, Wu W. Oral targeted drug delivery to post-gastrointestinal sites. J Control Release 2024; 370:256-276. [PMID: 38679163 DOI: 10.1016/j.jconrel.2024.04.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
As an essential branch of targeted drug delivery, oral targeted delivery is attracting growing attention in recent years. In addition to site-specific delivery for the treatment of locoregional diseases in the gastrointestinal tract (GIT), oral targeted delivery to remote sites beyond the GIT emerges as a cutting-edge research topic. This review aims to provide an overview of the fundamental concepts and most recent advances in this field. Owing to the physiological barriers existing in the GIT, carrier systems should be transported across the enteric epithelia to target remote sites. Recently, pioneer investigations have validated the transport of intact micro- or nanocarriers across gastrointestinal barriers and subsequently to various distal organs and tissues. The microfold (M) cell pathway is the leading mechanism underlying the oral absorption of particulates, but the contribution of the transcellular and paracellular pathways should not be neglected either. In addition to well-acknowledged physicochemical and biological factors, the formation of a protein corona may also influence the biological fate of carrier systems. Although in an early stage of conceptualization, oral targeted delivery to remote diseases has demonstrated promising potential for the treatment of inflammation, tumors, and diseases inflicting the lymphatic and mononuclear phagocytosis systems.
Collapse
Affiliation(s)
- Rongze Han
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China; Fudan Zhangjiang Institute, Shanghai 201203, China
| | - Huiping Lu
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Shun Shen
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China; Fudan Zhangjiang Institute, Shanghai 201203, China.
| |
Collapse
|
33
|
Saifi F, Jeoboam B, Demory Beckler M, Costin JM. The Association Between Lactational Infective Mastitis and the Microbiome: Development, Onset, and Treatments. Cureus 2024; 16:e62717. [PMID: 39036221 PMCID: PMC11259407 DOI: 10.7759/cureus.62717] [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: 03/15/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Lactational infective mastitis (LIM) was previously thought to occur due to trapped milk causing inadequate milk drainage and consequent infection. However, advances in genome sequencing techniques have shown that the abundance of Staphylococcus aureus, Staphylococcus epidermidis, Lactobacilli species, and Bifidobacterium species in the breast milk of lactating women play a key role in the development of LIM. Recent discoveries have revealed that the breast milk microbiome is composed of bacteria and other microorganisms, which are seeded through multiple pathways and are influenced by maternal factors. An imbalance in the microbial abundance in breast milk can lead to LIM. Given that this infection can cause early termination of breastfeeding, it is imperative to discuss prevention and treatment options. The objective of this review is to highlight the pathogens involved in LIM affecting human mothers, routes of bacterial transfer, and contributing factors that may influence changes in the composition of the milk microbiota, as well as propose preventative and curative treatment options.
Collapse
Affiliation(s)
- Farishta Saifi
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Benscar Jeoboam
- Biomedical Sciences, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Michelle Demory Beckler
- Microbiology and Immunology, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Joshua M Costin
- Medical Education, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
34
|
Del Castillo D, Lo DD. Deciphering the M-cell niche: insights from mouse models on how microfold cells "know" where they are needed. Front Immunol 2024; 15:1400739. [PMID: 38863701 PMCID: PMC11165056 DOI: 10.3389/fimmu.2024.1400739] [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: 03/14/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Known for their distinct antigen-sampling abilities, microfold cells, or M cells, have been well characterized in the gut and other mucosa including the lungs and nasal-associated lymphoid tissue (NALT). More recently, however, they have been identified in tissues where they were not initially suspected to reside, which raises the following question: what external and internal factors dictate differentiation toward this specific role? In this discussion, we will focus on murine studies to determine how these cells are identified (e.g., markers and function) and ask the broader question of factors triggering M-cell localization and patterning. Then, through the consideration of unconventional M cells, which include villous M cells, Type II taste cells, and medullary thymic epithelial M cells (microfold mTECs), we will establish the M cell as not just a player in mucosal immunity but as a versatile niche cell that adapts to its home tissue. To this end, we will consider the lymphoid structure relationship and apical stimuli to better discuss how the differing cellular programming and the physical environment within each tissue yield these cells and their unique organization. Thus, by exploring this constellation of M cells, we hope to better understand the multifaceted nature of this cell in its different anatomical locales.
Collapse
Affiliation(s)
| | - David D. Lo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
35
|
Jennings KC, Johnson KE, Hayward MA, Kristich CJ, Salzman NH. CCR2-dependent CX3CR1+ colonic macrophages promote Enterococcus faecalis dissemination. Infect Immun 2024; 92:e0000624. [PMID: 38629806 PMCID: PMC11075457 DOI: 10.1128/iai.00006-24] [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: 01/05/2024] [Accepted: 03/29/2024] [Indexed: 05/08/2024] Open
Abstract
Enterococci are common commensal bacteria that colonize the gastrointestinal tracts of most mammals, including humans. Importantly, these bacteria are one of the leading causes of nosocomial infections. This study examined the role of colonic macrophages in facilitating Enterococcus faecalis infections in mice. We determined that depletion of colonic phagocytes resulted in the reduction of E. faecalis dissemination to the gut-draining mesenteric lymph nodes. Furthermore, we established that trafficking of monocyte-derived CX3CR1-expressing macrophages contributed to E. faecalis dissemination in a manner that was not reliant on CCR7, the conventional receptor involved in lymphatic migration. Finally, we showed that E. faecalis mutants with impaired intracellular survival exhibited reduced dissemination, suggesting that E. faecalis can exploit host immune cell migration to disseminate systemically and cause disease. Our findings indicate that modulation of macrophage trafficking in the context of antibiotic therapy could serve as a novel approach for preventing or treating opportunistic infections by disseminating enteric pathobionts like E. faecalis.
Collapse
Affiliation(s)
- Kevin C. Jennings
- Department of Pediatrics, Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kaitlin E. Johnson
- Department of Pediatrics, Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael A. Hayward
- Department of Pediatrics, Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christopher J. Kristich
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Nita H. Salzman
- Department of Pediatrics, Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
36
|
Voges L, Weiß F, Branco AT, Fromm M, Krug SM. Expression and Localization Profiles of Tight Junction Proteins in Immune Cells Depend on Their Activation Status. Int J Mol Sci 2024; 25:4861. [PMID: 38732086 PMCID: PMC11084252 DOI: 10.3390/ijms25094861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
The ability of the immune system to combat pathogens relies on processes like antigen sampling by dendritic cells and macrophages migrating through endo- and epithelia or penetrating them with their dendrites. In addition, other immune cell subtypes also migrate through the epithelium after activation. For paracellular migration, interactions with tight junctions (TJs) are necessary, and previous studies reported TJ protein expression in several immune cells. Our investigation aimed to characterize, in more detail, the expression profiles of TJ proteins in different immune cells in both naïve and activated states. The mRNA expression analysis revealed distinct expression patterns for TJ proteins, with notable changes, mainly increases, upon activation. At the protein level, LSR appeared predominant, being constitutively present in naïve cell membranes, suggesting roles as a crucial interaction partner. Binding experiments suggested the presence of claudins in the membrane only after stimulation, and claudin-8 translocation to the membrane occurred after stimulation. Our findings suggest a dynamic TJ protein expression in immune cells, implicating diverse functions in response to stimulation, like interaction with TJ proteins or regulatory roles. While further analysis is needed to elucidate the precise roles of TJ proteins, our findings indicate important non-canonical functions of TJ proteins in immune response.
Collapse
Affiliation(s)
- Lena Voges
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Franziska Weiß
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Ana-Teresa Branco
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Michael Fromm
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Susanne M. Krug
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| |
Collapse
|
37
|
Marafini I, Monteleone I, Laudisi F, Monteleone G. Aryl Hydrocarbon Receptor Signalling in the Control of Gut Inflammation. Int J Mol Sci 2024; 25:4527. [PMID: 38674118 PMCID: PMC11050475 DOI: 10.3390/ijms25084527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Aryl hydrocarbon receptor (AHR), a transcription factor activated by many natural and synthetic ligands, represents an important mediator of the interplay between the environment and the host's immune responses. In a healthy gut, AHR activation promotes tolerogenic signals, which help maintain mucosal homeostasis. AHR expression is defective in the inflamed gut of patients with inflammatory bowel diseases (IBD), where decreased AHR signaling is supposed to contribute to amplifying the gut tissue's destructive immune-inflammatory responses. We here review the evidence supporting the role of AHR in controlling the "physiological" intestinal inflammation and summarize the data about the therapeutic effects of AHR activators, both in preclinical mouse models of colitis and in patients with IBD.
Collapse
Affiliation(s)
- Irene Marafini
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy;
| | - Ivan Monteleone
- Department of Biomedicine and Prevention, University of “Tor Vergata”, 00133 Rome, Italy;
| | - Federica Laudisi
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy;
| | - Giovanni Monteleone
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy;
- Department of Systems Medicine, University of “Tor Vergata”, 00133 Rome, Italy;
| |
Collapse
|
38
|
Dai Y, Zhang M, Liu X, Sun T, Qi W, Ding W, Chen Z, Zhang P, Liu R, Chen H, Chen S, Wang Y, Yue Y, Song N, Wang W, Jia H, Ma Z, Li C, Chen Q, Li B. Salmonella manipulates macrophage migration via SteC-mediated myosin light chain activation to penetrate the gut-vascular barrier. EMBO J 2024; 43:1499-1518. [PMID: 38528181 PMCID: PMC11021425 DOI: 10.1038/s44318-024-00076-7] [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: 07/30/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
The intestinal pathogen Salmonella enterica rapidly enters the bloodstream after the invasion of intestinal epithelial cells, but how Salmonella breaks through the gut-vascular barrier is largely unknown. Here, we report that Salmonella enters the bloodstream through intestinal CX3CR1+ macrophages during early infection. Mechanistically, Salmonella induces the migration/invasion properties of macrophages in a manner dependent on host cell actin and on the pathogen effector SteC. SteC recruits host myosin light chain protein Myl12a and phosphorylates its Ser19 and Thr20 residues. Myl12a phosphorylation results in actin rearrangement, and enhanced migration and invasion of macrophages. SteC is able to utilize a wide range of NTPs other than ATP to phosphorylate Myl12a. We further solved the crystal structure of SteC, which suggests an atypical dimerization-mediated catalytic mechanism. Finally, in vivo data show that SteC-mediated cytoskeleton manipulation is crucial for Salmonella breaching the gut vascular barrier and spreading to target organs.
Collapse
Affiliation(s)
- Yuanji Dai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Min Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoyu Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ting Sun
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wenqi Qi
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Ding
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhe Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Ping Zhang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ruirui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Huimin Chen
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Siyan Chen
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuzhen Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yingying Yue
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Nannan Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haihong Jia
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongrui Ma
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Cuiling Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
| | - Bingqing Li
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- School of Pharmaceutical Sciences, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
- Key Lab for Biotech-Drugs of National Health Commission, Jinan, 250117, China.
- Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, China.
| |
Collapse
|
39
|
Didriksen BJ, Eshleman EM, Alenghat T. Epithelial regulation of microbiota-immune cell dynamics. Mucosal Immunol 2024; 17:303-313. [PMID: 38428738 PMCID: PMC11412483 DOI: 10.1016/j.mucimm.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
The mammalian gastrointestinal tract hosts a diverse community of trillions of microorganisms, collectively termed the microbiota, which play a fundamental role in regulating tissue physiology and immunity. Recent studies have sought to dissect the cellular and molecular mechanisms mediating communication between the microbiota and host immune system. Epithelial cells line the intestine and form an initial barrier separating the microbiota from underlying immune cells, and disruption of epithelial function has been associated with various conditions ranging from infection to inflammatory bowel diseases and cancer. From several studies, it is now clear that epithelial cells integrate signals from commensal microbes. Importantly, these non-hematopoietic cells also direct regulatory mechanisms that instruct the recruitment and function of microbiota-sensitive immune cells. In this review, we discuss the central role that has emerged for epithelial cells in orchestrating intestinal immunity and highlight epithelial pathways through which the microbiota can calibrate tissue-intrinsic immune responses.
Collapse
Affiliation(s)
- Bailey J Didriksen
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily M Eshleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
40
|
Kaur J, Sharma A, Passi G, Dey P, Khajuria A, Alajangi HK, Jaiswal PK, Barnwal RP, Singh G. Nanomedicine at the Pulmonary Frontier: Immune-Centric Approaches for Respiratory Disease Treatment. Immunol Invest 2024; 53:295-347. [PMID: 38206610 DOI: 10.1080/08820139.2023.2298398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Respiratory diseases (RD) are a group of common ailments with a rapidly increasing global prevalence, posing a significant threat to humanity, especially the elderly population, and imposing a substantial burden on society and the economy. RD represents an unmet medical need that requires the development of viable pharmacotherapies. While various promising strategies have been devised to advance potential treatments for RD, their implementation has been hindered by difficulties in drug delivery, particularly in critically ill patients. Nanotechnology offers innovative solutions for delivering medications to the inflamed organ sites, such as the lungs. Although this approach is enticing, delivering nanomedicine to the lungs presents complex challenges that require sophisticated techniques. In this context, we review the potential of novel nanomedicine-based immunomodulatory strategies that could offer therapeutic benefits in managing this pressing health condition.
Collapse
Affiliation(s)
- Jatinder Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Akanksha Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Gautam Passi
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Piyush Dey
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Hema Kumari Alajangi
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, Texas, USA
| | | | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
41
|
Zhang Y, Zhao M, He J, Chen L, Wang W. In vitro and in vivo immunomodulatory activity of acetylated polysaccharides from Cyclocarya paliurus leaves. Int J Biol Macromol 2024; 259:129174. [PMID: 38181912 DOI: 10.1016/j.ijbiomac.2023.129174] [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: 04/24/2023] [Revised: 12/04/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
In this study, we aimed to investigate the immunomodulatory effects of polysaccharides from Cyclocarya paliurus leaves after acetylation modification (Ac-CPP0.1) on dendritic cells (DCs) and immunosuppressed mice. In vitro, Ac-CPP0.1 promoted phenotypic and functional maturation of DCs. Specifically, it increased the expression of costimulatory molecules (CD80, CD86, and MHC II) and the secretion of cytokines (TNF-α, IL-6, IL-1β, IL-10, IL-12p70) of DCs. In vivo, Ac-CPP0.1 significantly improved immunosuppression of mice, which was manifested by increased body weight and immune organ index, up-regulated cytokines (IL-4, IL-17, TGF-β3, and TNF-α), and restored short-chain fatty acid (SCFAs) levels of intestinal. The immunoactivation of Ac-CPP0.1 in DCs and in mice is linked to the activation of the TLR4/NF-κB signaling pathway. Furthermore, Ac-CPP0.1 reversed intestinal flora imbalance caused by cyclophosphamide. At the species level, Ac-CPP0.1 increased the abundance of unclassified_Muribaculaceae, unclassified_Desulfovibrio, Bacteroides_acidifaciens and Faecalibaculum_rodentium, decreased the level of Lactobacillus_johnsonii, unclassified_g_Staphylococcus and Staphylococcus_nepalensis. In summary, Ac-CPP0.1 has considerable immunomodulatory potential, which is beneficial to the future utilization and development of Cyclocarya paliurus.
Collapse
Affiliation(s)
- Yang Zhang
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Meng Zhao
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jing He
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Lingli Chen
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wenjun Wang
- Key Lab for Agro-product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
42
|
Pardy RD, Wallbank BA, Striepen B, Hunter CA. Immunity to Cryptosporidium: insights into principles of enteric responses to infection. Nat Rev Immunol 2024; 24:142-155. [PMID: 37697084 PMCID: PMC11881751 DOI: 10.1038/s41577-023-00932-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/13/2023]
Abstract
Cryptosporidium parasites replicate within intestinal epithelial cells and are an important cause of diarrhoeal disease in young children and in patients with primary and acquired defects in T cell function. This Review of immune-mediated control of Cryptosporidium highlights advances in understanding how intestinal epithelial cells detect this infection, the induction of innate resistance and the processes required for activation of T cell responses that promote parasite control. The development of a genetic tool set to modify Cryptosporidium combined with tractable mouse models provide new opportunities to understand the principles that govern the interface between intestinal epithelial cells and the immune system that mediate resistance to enteric pathogens.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
43
|
Grassi F, Marino R. The P2X7 receptor in mucosal adaptive immunity. Purinergic Signal 2024; 20:9-19. [PMID: 37067746 PMCID: PMC10828151 DOI: 10.1007/s11302-023-09939-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/31/2023] [Indexed: 04/18/2023] Open
Abstract
The P2X7 receptor (P2X7R) is a widely distributed cation channel activated by extracellular ATP (eATP) with exclusive peculiarities with respect to other P2XRs. In recent years, P2X7R has been shown to regulate the adaptive immune response by conditioning T cell signaling and activation as well as polarization, lineage stability, cell death, and function in tissues. Here we revise experimental observations in this field, with a focus on adaptive immunity at mucosal sites, particularly in the gut, where eATP is hypothesized to act in the reciprocal conditioning of the host immune system and commensal microbiota to promote mutualism. The importance of P2X7R activity in the intestine is consistent with the transcriptional upregulation of P2xr7 gene by retinoic acid, a metabolite playing a key role in mucosal immunity. We emphasize the function of the eATP/P2X7R axis in controlling T follicular helper (Tfh) cell in the gut-associated lymphoid tissue (GALT) and, consequently, T-dependent secretory IgA (SIgA), with a focus on high-affinity SIgA-mediated protection from enteropathogens and shaping of a beneficial microbiota for the host.
Collapse
Affiliation(s)
- Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università Della Svizzera Italiana, 6500, Bellinzona, Switzerland.
| | - Rebecca Marino
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università Della Svizzera Italiana, 6500, Bellinzona, Switzerland
| |
Collapse
|
44
|
Javan GT, Singh K, Finley SJ, Green RL, Sen CK. Complexity of human death: its physiological, transcriptomic, and microbiological implications. Front Microbiol 2024; 14:1345633. [PMID: 38282739 PMCID: PMC10822681 DOI: 10.3389/fmicb.2023.1345633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Human death is a complex, time-governed phenomenon that leads to the irreversible cessation of all bodily functions. Recent molecular and genetic studies have revealed remarkable experimental evidence of genetically programmed cellular death characterized by several physiological processes; however, the basic physiological function that occurs during the immediate postmortem period remains inadequately described. There is a paucity of knowledge connecting necrotic pathologies occurring in human organ tissues to complete functional loss of the human organism. Cells, tissues, organs, and organ systems show a range of differential resilience and endurance responses that occur during organismal death. Intriguingly, a persistent ambiguity in the study of postmortem physiological systems is the determination of the trajectory of a complex multicellular human body, far from life-sustaining homeostasis, following the gradual or sudden expiry of its regulatory systems. Recent groundbreaking investigations have resulted in a paradigm shift in understanding the cell biology and physiology of death. Two significant findings are that (i) most cells in the human body are microbial, and (ii) microbial cell abundance significantly increases after death. By addressing the physiological as well as the microbiological aspects of death, future investigations are poised to reveal innovative insights into the enigmatic biological activities associated with death and human decomposition. Understanding the elaborate crosstalk of abiotic and biotic factors in the context of death has implications for scientific discoveries important to informing translational knowledge regarding the transition from living to the non-living. There are important and practical needs for a transformative reestablishment of accepted models of biological death (i.e., artificial intelligence, AI) for more precise determinations of when the regulatory mechanisms for homeostasis of a living individual have ceased. In this review, we summarize mechanisms of physiological, genetic, and microbiological processes that define the biological changes and pathways associated with human organismal death and decomposition.
Collapse
Affiliation(s)
- Gulnaz T. Javan
- Department of Physical and Forensic Sciences, Alabama State University, Montgomery, AL, United States
| | - Kanhaiya Singh
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sheree J. Finley
- Department of Physical and Forensic Sciences, Alabama State University, Montgomery, AL, United States
| | - Robert L. Green
- Department of Physical and Forensic Sciences, Alabama State University, Montgomery, AL, United States
| | - Chandan K. Sen
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
45
|
Xu S, Chen S, Liu Y, Jia X, Jiang X, Che L, Lin Y, Zhuo Y, Feng B, Fang Z, Li J, Hua L, Wang J, Zhang R, Ren Z, Wu D. Generation of Porcine Angiogenin 4-Expressing Pichia pastoris and Its Protection against Intestinal Inflammatory Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:153-165. [PMID: 38130066 DOI: 10.1021/acs.jafc.3c05789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Antimicrobial peptides have been extensively studied as potential alternatives to antibiotics. Porcine angiogenin 4 (pANG4) is a novel antimicrobial peptide in the angiogenin (ANG) family, which may have a regulatory effect on intestinal microflora. The object of present study is obtained pANG4 protein by heterologous expression, so as to explore the biological function of recombinant pANG4 (rpANG4). The pANG4 was expressed in Pichia pastoris (P. pastoris) and anti-inflammatory effects were investigated in intestinal porcine epithelial cell line-J2 (IPEC-J2) and mice. Purified rpANG4 had bacteriostatic activity and did not cause hemolysis or cytotoxicity at concentrations below 128 μg/mL. Purified rpANG4 increased the activity of IPEC-J2 and reduced apoptosis in vitro. rpANG4 reduced the pro-inflammatory gene expression and upregulated tight junction protein gene expression during inflammation. rpANG4 alleviated lipopolysaccharide (LPS)-induced liver and spleen damage, intestinal inflammation, jejunal apoptosis genes' expression, and improved immune function in an in vivo mice model. rpANG4 increased tight junction protein gene expression in jejunum, thereby improving the jejunum intestinal barrier function. In conclusion, rpANG4 had antibacterial activity, inhibited intestinal inflammation, improved intestinal barrier function, and alleviated liver and spleen damage. The current study contributes to the development of antibiotic substitutes and the improvement of animal health.
Collapse
Affiliation(s)
- Shengyu Xu
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Sirun Chen
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Yalei Liu
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Xinlin Jia
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Xuemei Jiang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Lianqiang Che
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Yan Lin
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Yong Zhuo
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Bin Feng
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Zhengfeng Fang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Jian Li
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Lun Hua
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Jianping Wang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Ruinan Zhang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Province Key Laboratory of Animal Disease and Human Health, Key Laboratory of Environmental Hazard and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, P. R. China
| | - De Wu
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, P. R. China
| |
Collapse
|
46
|
He J, Cao Y, Zhu Q, Wang X, Cheng G, Wang Q, He R, Lu H, Weng Y, Mao G, Bao Y, Wang J, Liu X, Han F, Shi P, Shen XZ. Renal macrophages monitor and remove particles from urine to prevent tubule obstruction. Immunity 2024; 57:106-123.e7. [PMID: 38159573 DOI: 10.1016/j.immuni.2023.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/17/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024]
Abstract
When the filtrate of the glomerulus flows through the renal tubular system, various microscopic sediment particles, including mineral crystals, are generated. Dislodging these particles is critical to ensuring the free flow of filtrate, whereas failure to remove them will result in kidney stone formation and obstruction. However, the underlying mechanism for the clearance is unclear. Here, using high-resolution microscopy, we found that the juxtatubular macrophages in the renal medulla constitutively formed transepithelial protrusions and "sampled" urine contents. They efficiently sequestered and phagocytosed intraluminal sediment particles and occasionally transmigrated to the tubule lumen to escort the excretion of urine particles. Mice with decreased renal macrophage numbers were prone to developing various intratubular sediments, including kidney stones. Mechanistically, the transepithelial behaviors of medulla macrophages required integrin β1-mediated ligation to the tubular epithelium. These findings indicate that medulla macrophages sample urine content and remove intratubular particles to keep the tubular system unobstructed.
Collapse
Affiliation(s)
- Jian He
- Department of Physiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yangyang Cao
- Department of Physiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qian Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinge Wang
- Department of Physiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guo Cheng
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Wang
- Department of Laboratory Medicine, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Rukun He
- Department of Physiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haoran Lu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, Zhejiang, China
| | - Yuancheng Weng
- Department of Physiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yizhong Bao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Liu
- Department of Neurology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fei Han
- Kidney Disease Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng Shi
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xiao Z Shen
- Department of Physiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
47
|
Ichikawa M, Okada H, Nakamoto N, Taniki N, Chu PS, Kanai T. The gut-liver axis in hepatobiliary diseases. Inflamm Regen 2024; 44:2. [PMID: 38191517 PMCID: PMC10773109 DOI: 10.1186/s41232-023-00315-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/17/2023] [Indexed: 01/10/2024] Open
Abstract
Recent advances in the analysis of intestinal bacteria have led to reports of variations in intestinal bacterial levels among hepatobiliary diseases. The mechanisms behind the changes in intestinal bacteria in various hepatobiliary diseases include the abnormal composition of intestinal bacteria, weakening of the intestinal barrier, and bacterial translocation outside the intestinal tract, along with their metabolites, but many aspects remain unresolved. Further research employing clinical studies and animal models is expected to clarify the direct relationship between intestinal bacteria and hepatobiliary diseases and to validate the utility of intestinal bacteria as a diagnostic biomarker and potential therapeutic target. This review summarizes the involvement of the microbiota in the pathogenesis of hepatobiliary diseases via the gut-liver axis.
Collapse
Affiliation(s)
- Masataka Ichikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Haruka Okada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan.
| | - Nobuhito Taniki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Po-Sung Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, 1608582, Japan.
| |
Collapse
|
48
|
McDaniel ZS, Hales KE, Salih H, Deters A, Shi X, Nagaraja TG, Lawrence TE, Tennant TC, Amachawadi RG, Carroll JA, Burdick Sanchez NC, Galyean ML, Smock TM, Ballou MA, Machado VS, Davis E, Broadway PR. Development of an experimental model for liver abscess induction in Holstein steers using an acidotic diet challenge and bacterial inoculation. J Anim Sci 2024; 102:skae046. [PMID: 38447078 PMCID: PMC10941643 DOI: 10.1093/jas/skae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/04/2024] [Indexed: 03/08/2024] Open
Abstract
Holstein steers (n = 40; initial BW = 84.9 ± 7.1 kg) were used to study the genesis of liver abscesses (LA) using an acidotic diet challenge with or without intraruminal bacterial inoculation. Steers were housed in individual pens inside a barn and randomly assigned to one of three treatments: (1) low-starch control diet comprised primarily of dry-rolled corn and wet corn gluten feed (CON); (2) high-starch acidotic diet with steam-flaked corn (AD); or (3) acidotic diet plus intraruminal inoculation with Fusobacterium necrophorum subsp. necrophorum (9.8 × 108 colony forming units [CFU]/mL), Trueperella pyogenes (3.91 × 109 CFU/mL), and Salmonella enterica serovar Lubbock (3.07 × 108 CFU/mL), previously isolated from LA (ADB). Steers in AD and ADB were fed the acidotic diet for 3 d followed by 2 d of the CON diet, and this cycle was repeated four times. On day 23, ADB steers were intraruminally inoculated with the bacteria. At necropsy, gross pathology of livers, lungs, rumens, and colons was noted. Continuous data were analyzed via mixed models as repeated measures over time with individual steer as the experimental unit. Mixed models were also used to determine the difference in prevalence of necropsy scores among treatments. Ruminal pH decreased in AD and ADB steers during each acidotic diet cycle (P ≤ 0.05). LA prevalence was 42.9% (6 of 14) in ADB vs. 0% in AD or CON treatments (P < 0.01). Ruminal damage was 51.1% greater in ADB than in AD (P ≤ 0.04). Culture of LA determined that 100% of the abscesses contained F. necrophorum subsp. necrophorum, 0% contained T. pyogenes, 50% contained Salmonella, and 50% contained a combination of F. necrophorum subsp. necrophorum and Salmonella. The F. necrophorum subsp. necrophorum was clonally identical to the strain used for the bacterial inoculation based on phylogenetic analysis of the whole genome. This experimental model successfully induced rumenitis and LA in Holstein steers and confirms the central dogma of LA pathogenesis that acidosis and rumenitis lead to the entry of F. necrophorum into the liver to cause abscesses. Our findings suggest that an acidotic diet, in conjunction with intraruminal bacterial inoculation, is a viable model to induce LA. Further research is needed to determine the repeatability of this model, and a major application of the model will be in evaluations of novel interventions to prevent LA.
Collapse
Affiliation(s)
- Zach S McDaniel
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX, USA
| | - Kristin E Hales
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX, USA
| | - Harith Salih
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Alyssa Deters
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Xiaorong Shi
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | | | - Ty E Lawrence
- Department of Agricultural Sciences, West Texas A&M University, Canyon, TX, USA
| | - Travis C Tennant
- Department of Agricultural Sciences, West Texas A&M University, Canyon, TX, USA
| | | | - Jeff A Carroll
- United States Department of Agriculture, Agricultural Research Service, Livestock Issues Research Unit, Lubbock, TX, USA
| | - Nicole C Burdick Sanchez
- United States Department of Agriculture, Agricultural Research Service, Livestock Issues Research Unit, Lubbock, TX, USA
| | - Michael L Galyean
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX, USA
| | - Taylor M Smock
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX, USA
| | - Michael A Ballou
- Department of Veterinary Sciences, Texas Tech University, Lubbock, TX, USA
| | - Vinicius S Machado
- Department of Veterinary Sciences, Texas Tech University, Lubbock, TX, USA
| | - Emily Davis
- Department of Veterinary Sciences, Texas Tech University, Lubbock, TX, USA
| | - Paul R Broadway
- United States Department of Agriculture, Agricultural Research Service, Livestock Issues Research Unit, Lubbock, TX, USA
| |
Collapse
|
49
|
Valdebenito S, Ajasin D, Valerdi K, Liu YR, Rao S, Eugenin EA. Mechanisms of Intracellular Communication in Cancer and Pathogen Spreading. Results Probl Cell Differ 2024; 73:301-326. [PMID: 39242384 DOI: 10.1007/978-3-031-62036-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Cell-to-cell interactions are essential for proper development, homeostasis, and complex syncytia/organ formation and function. Intercellular communication are mediated by multiple mechanisms including soluble mediators, adhesion molecules and specific mechanisms of cell to cell communication such as Gap junctions (GJ), tunneling nanotubes (TNT), and exosomes. Only recently, has been discovered that TNTs and exosomes enable the exchange of large signaling molecules, RNA, viral products, antigens, and organelles opening new avenues of research and therapeutic approaches. The focus of this review is to summarize these recent findings in physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - David Ajasin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Karl Valerdi
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | | | - Samvrit Rao
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, USA
| | - Eliseo A Eugenin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA.
| |
Collapse
|
50
|
Wheeler R, Gomperts Boneca I. The hidden base of the iceberg: gut peptidoglycome dynamics is foundational to its influence on the host. Gut Microbes 2024; 16:2395099. [PMID: 39239828 PMCID: PMC11382707 DOI: 10.1080/19490976.2024.2395099] [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: 12/21/2023] [Revised: 07/01/2024] [Accepted: 08/16/2024] [Indexed: 09/07/2024] Open
Abstract
The intestinal microbiota of humans includes a highly diverse range of bacterial species. All these bacteria possess a cell wall, composed primarily of the macromolecule peptidoglycan. As such, the gut also harbors an abundant and varied peptidoglycome. A remarkable range of host physiological pathways are regulated by peptidoglycan fragments that originate from the gut microbiota and enter the host system. Interactions between the host system and peptidoglycan can influence physiological development and homeostasis, promote health, or contribute to inflammatory disease. Underlying these effects is the interplay between microbiota composition and enzymatic processes that shape the intestinal peptidoglycome, dictating the types of peptidoglycan generated, that subsequently cross the gut barrier. In this review, we highlight and discuss the hidden and emerging functional aspects of the microbiome, i.e. the hidden base of the iceberg, that modulate the composition of gut peptidoglycan, and how these fundamental processes are drivers of physiological outcomes for the host.
Collapse
Affiliation(s)
- Richard Wheeler
- Institut Pasteur, Université Paris Cité, Paris, France
- Hauts-de-Seine, Arthritis Research and Development, Neuilly-sur-Seine, France
| | | |
Collapse
|