1
|
Zhou Z, Su J, van Os BW, Plug LG, de Jonge-Muller ESM, Brands L, Janson SGT, van de Beek LM, van der Meulen-de Jong AE, Hawinkels LJAC, Barnhoorn MC. Stromal Cell Subsets Show Model-Dependent Changes in Experimental Colitis and Affect Epithelial Tissue Repair and Immune Cell Activation. Inflamm Bowel Dis 2025; 31:1051-1066. [PMID: 40100003 PMCID: PMC11985400 DOI: 10.1093/ibd/izae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Indexed: 03/20/2025]
Abstract
BACKGROUND Previous work on inflammatory bowel disease (IBD) revealed changes in the abundance of colonic stromal subsets during intestinal inflammation. However, it is currently unknown whether these stromal cell subset changes are also reflected in different IBD mouse models and how commonly used IBD therapies affect stromal cell subset composition. METHODS Stromal subset markers CD55, C-X-C motif chemokine 12 (CXCL12), podoplanin (PDPN), CD90, and CD73 were analyzed by flow cytometry in 3 mouse models for IBD, namely interleukin (IL)-10 knockout (KO), dextran sulfate sodium-induced, and T-cell transfer model for colitis. Next, the effects of IBD therapies on the stromal subset composition were studied. In vitro experiments were performed to study the interaction between stromal cell subsets and epithelial/immune cells. RESULTS The colitis-induced changes in the abundance of stromal cell subsets differed considerably between the 3 colitis mouse models. Interestingly, treatment with IBD medication affected specific stromal subsets in a therapy and model-specific manner. In vitro experiments showed that specific stromal subsets affected epithelial wound healing and/or T-cell activation. CONCLUSIONS The relative abundance changes of stromal cell subsets during experimental colitis differ between 3 established colitis models. Treatment with IBD therapies influences stromal subset abundance, indicating their importance in IBD pathogenesis, possibly through affecting epithelial migration, and T-cell activation.
Collapse
Affiliation(s)
- Zhou Zhou
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Jie Su
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bram W van Os
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Leonie G Plug
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | | | - Stef G T Janson
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | - Andrea E van der Meulen-de Jong
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Marieke C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
2
|
Kreimeyer H, Llorente C, Schnabl B. Influence of Alcohol on the Intestinal Immune System. Alcohol Res 2025; 45:03. [PMID: 40151622 PMCID: PMC11913448 DOI: 10.35946/arcr.v45.1.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Alcohol misuse is associated with disruption of the microbial homeostasis (dysbiosis) and microbial overgrowth in the gut, gut barrier disruption, and translocation of microbes into the systemic circulation. It also induces changes in regulatory mechanisms of the gut, which is the largest peripheral immune organ. The gut-liver axis is important for health and disease, and alterations in the intestinal immune system contribute to alcohol-associated liver disease (ALD). Understanding these changes might help discover new targets for drugs and therapeutic approaches. SEARCH METHODS A systematic literature search was conducted in PubMed, Medline, and Embase of manuscripts published between January 2000 and November 2023 using the terms ("alcohol" or "ethanol") AND ("immune" or "immunol") AND ("intestine," "colon," or "gut"). Eligible manuscripts included studies and reviews that discussed the effects of ethanol on immune cells in the intestine. SEARCH RESULTS A total of 506 publications were found in the databases on November 20, 2023. After excluding duplicates and research not covering ALD (415 articles), 91 studies were reviewed. Also included were manuscripts covering specific immune cells in the context of ALD. DISCUSSION AND CONCLUSIONS Balancing immune tolerance vs. initiating an immune response challenges the intestinal immune system. Alcohol induces disruption of the intestinal barrier, which is accompanied by a thicker mucus layer and reduced anti-microbial peptides. This leads to longer attachment of bacteria to epithelial cells and consequently greater translocation into the circulation. Bacterial translocation activates the immune system, reducing the activity of regulatory T cells and inducing T helper 17 response via a variety of pathways. The role of innate immune cells, especially Type 3 innate lymphoid cells, and of specific B- and T-cell subsets in ALD remains elusive. Gut dysbiosis, translocation of viable bacteria and bacterial products into the circulation, and changes in the intestinal barrier have been linked to immune deficiency and infections in patients with cirrhosis. Modifying the intestinal immune system could reduce intestinal inflammation and alcohol-induced liver injury. Understanding the underlying pathophysiology can help to detect new targets for drugs and design therapeutic strategies.
Collapse
Affiliation(s)
- Henriette Kreimeyer
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California
- Department of Medicine, U.S. Department of Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
3
|
Özkan A, LoGrande NT, Feitor JF, Goyal G, Ingber DE. Intestinal organ chips for disease modelling and personalized medicine. Nat Rev Gastroenterol Hepatol 2024; 21:751-773. [PMID: 39192055 DOI: 10.1038/s41575-024-00968-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/29/2024]
Abstract
Alterations in intestinal structure, mechanics and physiology underlie acute and chronic intestinal conditions, many of which are influenced by dysregulation of microbiome, peristalsis, stroma or immune responses. Studying human intestinal physiology or pathophysiology is difficult in preclinical animal models because their microbiomes and immune systems differ from those of humans. Although advances in organoid culture partially overcome this challenge, intestinal organoids still lack crucial features that are necessary to study functions central to intestinal health and disease, such as digestion or fluid flow, as well as contributions from long-term effects of living microbiome, peristalsis and immune cells. Here, we review developments in organ-on-a-chip (organ chip) microfluidic culture models of the human intestine that are lined by epithelial cells and interfaced with other tissues (such as stroma or endothelium), which can experience both fluid flow and peristalsis-like motions. Organ chips offer powerful ways to model intestinal physiology and disease states for various human populations and individual patients, and can be used to gain new insight into underlying molecular and biophysical mechanisms of disease. They can also be used as preclinical tools to discover new drugs and then validate their therapeutic efficacy and safety in the same human-relevant model.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Nina Teresa LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jessica F Feitor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
4
|
Dang EV, Reboldi A. Cholesterol sensing and metabolic adaptation in tissue immunity. Trends Immunol 2024; 45:861-870. [PMID: 39424470 PMCID: PMC11560508 DOI: 10.1016/j.it.2024.09.013] [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: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Cholesterol metabolites, particularly oxidized forms known as oxysterols, play crucial roles in modulating immune and metabolic processes across various tissues. Concentrations of local cholesterol and its metabolites influence tissue-specific immune responses by shaping the metabolic and spatial organization of immune cells in barrier organs like the small intestine (SI) and lungs. We explore recent molecular and cellular evidence supporting the metabolic adaptation of innate and adaptive immune cells in the SI and lung, driven by cholesterol and cholesterol metabolites. Further research should unravel the detailed molecular mechanisms and spatiotemporal adaptations involving cholesterol metabolites in distinct mucosal tissues in homeostasis or infection. We posit that pharmacological interventions targeting the generation or sensing of cholesterol metabolites might be leveraged to enhance long-term immune protection in mucosal tissues or prevent autoinflammatory states.
Collapse
Affiliation(s)
- Eric V Dang
- Molecular Mycology and Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Andrea Reboldi
- Department of Pathology, Immunology, and Microbial Pathogenesis Program, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
5
|
Yokoi F, Deguchi S, Watanabe Y, Takayama K. Establishment of an ulcerative colitis model using colon organoids derived from human induced pluripotent stem cells. iScience 2024; 27:111049. [PMID: 39435148 PMCID: PMC11492162 DOI: 10.1016/j.isci.2024.111049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/31/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
The etiology of inflammatory bowel disease (IBD) is complex, with much room for a greater understanding and development of improved therapies. Therefore, establishing a reliable IBD model is crucial for future advancements. In this study, human induced pluripotent stem (iPS) cell-derived colon organoids (hiPSC-COs) were treated with a combination of tumor necrosis factor alpha (TNF-α), interferon-gamma (IFN-γ), and interleukin (IL)-1β (3 cytokines [3CK]), known to be elevated in the serum of IBD patients. Inflammatory responses in stromal cells and damage to intestinal epithelial cells were observed in the 3CK-treated hiPSC-COs. Comparison of molecular signatures of 3CK-treated hiPSC-COs with those of ulcerative colitis (UC) patient's colon revealed that 3CK-treated hiPSC-COs resemble UC patient's colon. Furthermore, the elevated production of inflammatory cytokines observed in 3CK-treated hiPSC-COs was attenuated by treatment with tofacitinib. Our UC model will be an essential tool to understand its pathologic mechanisms and identify effective therapeutic approaches.
Collapse
Affiliation(s)
- Fuki Yokoi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yukio Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| |
Collapse
|
6
|
Smythies LE, Belyaeva OV, Alexander KL, Bimczok D, Nick HJ, Serrano CA, Huff KR, Nearing M, Musgrove L, Poovey EH, Garth J, Russ K, Baig KRKK, Crossman DK, Peter S, Cannon JA, Elson CO, Kedishvili NY, Smith PD. Human intestinal stromal cells promote homeostasis in normal mucosa but inflammation in Crohn's disease in a retinoic acid-deficient manner. Mucosal Immunol 2024; 17:958-972. [PMID: 38945396 PMCID: PMC11530961 DOI: 10.1016/j.mucimm.2024.06.009] [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: 02/13/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
Intestinal stromal cells (SCs), which synthesize the extracellular matrix that gives the mucosa its structure, are newly appreciated to play a role in mucosal inflammation. Here, we show that human intestinal vimentin+CD90+smooth muscle actin- SCs synthesize retinoic acid (RA) at levels equivalent to intestinal epithelial cells, a function in the human intestine previously attributed exclusively to epithelial cells. Crohn's disease SCs (Crohn's SCs), however, synthesized markedly less RA than SCs from healthy intestine (normal SCs). We also show that microbe-stimulated Crohn's SCs, which are more inflammatory than stimulated normal SCs, induced less RA-regulated differentiation of mucosal dendritic cells (DCs) (circulating pre-DCs and monocyte-derived DCs), leading to the generation of more potent inflammatory interferon-γhi/interleukin-17hi T cells than normal SCs. Explaining these results, Crohn's SCs expressed more DHRS3, a retinaldehyde reductase that inhibits retinol conversion to retinal and, thus, synthesized less RA than normal SCs. These findings uncover a microbe-SC-DC crosstalk in which luminal microbes induce Crohn's disease SCs to initiate and perpetuate inflammation through impaired synthesis of RA.
Collapse
Affiliation(s)
- Lesley E Smythies
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Olga V Belyaeva
- Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Katie L Alexander
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Diane Bimczok
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Heidi J Nick
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Carolina A Serrano
- Department of Pediatric Gastroenterology and Nutrition, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Kayci R Huff
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marie Nearing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lois Musgrove
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily H Poovey
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jaleesa Garth
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kirk Russ
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kondal R K K Baig
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shajan Peter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jamie A Cannon
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles O Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Natalia Y Kedishvili
- Department of Biochemistry and Molecular Genetics, Schools of Medicine and Dentistry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Phillip D Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
González A, Fullaondo A, Odriozola A. Microbiota-associated mechanisms in colorectal cancer. ADVANCES IN GENETICS 2024; 112:123-205. [PMID: 39396836 DOI: 10.1016/bs.adgen.2024.05.002] [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: 10/15/2024]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, ranking third in terms of incidence and second as a cause of cancer-related death. There is growing scientific evidence that the gut microbiota plays a key role in the initiation and development of CRC. Specific bacterial species and complex microbial communities contribute directly to CRC pathogenesis by promoting the neoplastic transformation of intestinal epithelial cells or indirectly through their interaction with the host immune system. As a result, a protumoural and immunosuppressive environment is created conducive to CRC development. On the other hand, certain bacteria in the gut microbiota contribute to protection against CRC. In this chapter, we analysed the relationship of the gut microbiota to CRC and the associations identified with specific bacteria. Microbiota plays a key role in CRC through various mechanisms, such as increased intestinal permeability, inflammation and immune system dysregulation, biofilm formation, genotoxin production, virulence factors and oxidative stress. Exploring the interaction between gut microbiota and tumourigenesis is essential for developing innovative therapeutic approaches in the fight against CRC.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| |
Collapse
|
8
|
Lassoued N, Yero A, Jenabian MA, Soret R, Pilon N. Efficient enzyme-free method to assess the development and maturation of the innate and adaptive immune systems in the mouse colon. Sci Rep 2024; 14:11063. [PMID: 38744932 PMCID: PMC11094196 DOI: 10.1038/s41598-024-61834-5] [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: 01/08/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
Researchers who aim to globally analyze the gastrointestinal immune system via flow cytometry have many protocol options to choose from, with specifics generally tied to gut wall layers of interest. To get a clearer idea of the approach we should use on full-thickness colon samples from mice, we first undertook a systematic comparison of three tissue dissociation techniques: two based on enzymatic cocktails and the other one based on manual crushing. Using flow cytometry panels of general markers of lymphoid and myeloid cells, we found that the presence of cell-surface markers and relative cell population frequencies were more stable with the mechanical method. Both enzymatic approaches were associated with a marked decrease of several cell-surface markers. Using mechanical dissociation, we then developed two minimally overlapping panels, consisting of a total of 26 antibodies, for serial profiling of lymphoid and myeloid lineages from the mouse colon in greater detail. Here, we highlight how we accurately delineate these populations by manual gating, as well as the reproducibility of our panels on mouse spleen and whole blood. As a proof-of-principle of the usefulness of our general approach, we also report segment- and life stage-specific patterns of immune cell profiles in the colon. Overall, our data indicate that mechanical dissociation is more suitable and efficient than enzymatic methods for recovering immune cells from all colon layers at once. Additionally, our panels will provide researchers with a relatively simple tool for detailed immune cell profiling in the murine gastrointestinal tract, regardless of life stage or experimental conditions.
Collapse
Affiliation(s)
- Nejia Lassoued
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
| | - Alexis Yero
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada
- Human Immuno-Virology Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Rodolphe Soret
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada.
- Centre d'excellence en recherche sur les maladies orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC, Canada.
- Department of Pediatrics, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
9
|
Asal M, Rep M, Bontkes HJ, van Vliet SJ, Mebius RE, Gibbs S. Towards Full Thickness Small Intestinal Models: Incorporation of Stromal Cells. Tissue Eng Regen Med 2024; 21:369-377. [PMID: 38113015 PMCID: PMC10987430 DOI: 10.1007/s13770-023-00600-6] [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: 06/02/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 12/21/2023] Open
Abstract
INTRODUCTION Since small intestine is one of the major barriers of the human body, there is a need to develop reliable in vitro human small intestinal models. These models should incorporate both the epithelial and lamina propria compartments and have similar barrier properties compared to that of the human tissue. These properties are essential for various applications, such as studying cell-cell interaction, intestinal diseases and testing permeability and metabolism of drugs and other compounds. The small intestinal lamina propria contains multiple stromal cell populations with several important functions, such as secretion of extracellular matrix proteins and soluble mediators. In addition, stromal cells influence the intestinal epithelial barrier, support the intestinal stem cell niche and interact with immune cells. METHODS In this review, we provide an extensive overview on the different types of lamina propria stromal cells found in small intestine and describe a combination of molecular markers that can be used to distinguish each different stromal cell type. We focus on studies that incorporated stromal cells into human representative small intestine models cultured on transwells. RESULTS AND CONCLUSION These models display enhanced epithelial morphology, increased cell proliferation and human-like barrier properties, such as low transepithelial electrical resistance (TEER) and intermediate permeability, thus better mimicking the native human small intestine than models only consisting of an epithelium which generally show high TEER and low permeability.
Collapse
Affiliation(s)
- Melis Asal
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Mila Rep
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Hetty J Bontkes
- Laboratory Medical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Sandra J van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands.
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Ventrello SW, McMurry NR, Edwards NM, Bain LJ. Chronic arsenic exposure affects stromal cells and signaling in the small intestine in a sex-specific manner. Toxicol Sci 2024; 198:303-315. [PMID: 38310360 PMCID: PMC10964740 DOI: 10.1093/toxsci/kfae016] [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: 02/05/2024] Open
Abstract
Arsenic is a toxicant that is ingested through drinking water and food, exposing nearly 140 million people to levels above the 10 ppb guideline concentration. Studies have shown that arsenic affects intestinal stem cells (ISCs), but the mechanisms by which arsenic alters the formation of adult cells in the small intestine are not well understood. Signals derived from intestinal stromal cells initiate and maintain differentiation. The goal of this study is to evaluate arsenic's effect on intestinal stromal cells, including PdgfrαLo trophocytes, located proximal to the ISCs, and PdgfrαHi telocytes, located proximal to the transit-amplifying region and up the villi. Adult Sox9tm2Crm-EGFP mice were exposed to 0, 33, and 100 ppb sodium arsenite in their drinking water for 13 weeks, and sections of duodenum were examined. Flow cytometry indicated that arsenic exposure dose-responsively reduced Sox9+ epithelial cells and trended toward increased Pdgfrα+ cells. The trophocyte marker, CD81, was reduced by 10-fold and 9.0-fold in the 100 ppb exposure group in male and female mice, respectively. Additionally, a significant 2.2- to 3.1-fold increase in PdgfrαLo expression was found in male mice in trophocytes and Igfbp5+ cells. PdgfrαHi protein expression, a telocyte marker, was more prevalent along the villus/crypt structure in females, whereas Gli1 expression (telocytes) was reduced in male mice exposed to arsenic. Principle coordinate analysis confirmed the sex-dependent response to arsenic exposure, with an increase in trophocyte and decrease in telocyte marker expression observed in male mice. These results imply that arsenic alters intestinal mesenchymal cells in a sex-dependent manner.
Collapse
Affiliation(s)
- Scott W Ventrello
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas R McMurry
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Nicholas M Edwards
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634, USA
| |
Collapse
|
11
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
12
|
El Tekle G, Andreeva N, Garrett WS. The Role of the Microbiome in the Etiopathogenesis of Colon Cancer. Annu Rev Physiol 2024; 86:453-478. [PMID: 38345904 DOI: 10.1146/annurev-physiol-042022-025619] [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: 02/15/2024]
Abstract
Studies in preclinical models support that the gut microbiota play a critical role in the development and progression of colorectal cancer (CRC). Specific microbial species and their corresponding virulence factors or associated small molecules can contribute to CRC development and progression either via direct effects on the neoplastic transformation of epithelial cells or through interactions with the host immune system. Induction of DNA damage, activation of Wnt/β-catenin and NF-κB proinflammatory pathways, and alteration of the nutrient's availability and the metabolic activity of cancer cells are the main mechanisms by which the microbiota contribute to CRC. Within the tumor microenvironment, the gut microbiota alter the recruitment, activation, and function of various immune cells, such as T cells, macrophages, and dendritic cells. Additionally, the microbiota shape the function and composition of cancer-associated fibroblasts and extracellular matrix components, fashioning an immunosuppressive and pro-tumorigenic niche for CRC. Understanding the complex interplay between gut microbiota and tumorigenesis can provide therapeutic opportunities for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Geniver El Tekle
- Department of Immunology and Infectious Diseases and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA;
- The Harvard Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Natalia Andreeva
- Department of Immunology and Infectious Diseases and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA;
- The Harvard Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA;
- The Harvard Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Caetano A, Sharpe P. Redefining Mucosal Inflammation with Spatial Genomics. J Dent Res 2024; 103:129-137. [PMID: 38166489 PMCID: PMC10845836 DOI: 10.1177/00220345231216114] [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: 01/04/2024] Open
Abstract
The human oral mucosa contains one of the most complex cellular systems that are essential for normal physiology and defense against a wide variety of local pathogens. Evolving techniques and experimental systems have helped refine our understanding of this complex cellular network. Current single-cell RNA sequencing methods can resolve subtle differences between cell types and states, thus providing a great tool for studying the molecular and cellular repertoire of the oral mucosa in health and disease. However, it requires the dissociation of tissue samples, which means that the interrelationships between cells are lost. Spatial transcriptomic methods bypass tissue dissociation and retain this spatial information, thereby allowing gene expression to be assessed across thousands of cells within the context of tissue structural organization. Here, we discuss the contribution of spatial technologies in shaping our understanding of this complex system. We consider the impact on identifying disease cellular neighborhoods and how space defines cell state. We also discuss the limitations and future directions of spatial sequencing technologies with recent advances in machine learning. Finally, we offer a perspective on open questions about mucosal homeostasis that these technologies are well placed to address.
Collapse
Affiliation(s)
- A.J. Caetano
- Centre for Oral Immunobiology and Regenerative Medicine, Barts Centre for Squamous Cancer, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - P.T. Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London, UK
| |
Collapse
|
14
|
Cameron O, Neves JF, Gentleman E. Listen to Your Gut: Key Concepts for Bioengineering Advanced Models of the Intestine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302165. [PMID: 38009508 PMCID: PMC10837392 DOI: 10.1002/advs.202302165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/12/2023] [Indexed: 11/29/2023]
Abstract
The intestine performs functions central to human health by breaking down food and absorbing nutrients while maintaining a selective barrier against the intestinal microbiome. Key to this barrier function are the combined efforts of lumen-lining specialized intestinal epithelial cells, and the supportive underlying immune cell-rich stromal tissue. The discovery that the intestinal epithelium can be reproduced in vitro as intestinal organoids introduced a new way to understand intestinal development, homeostasis, and disease. However, organoids reflect the intestinal epithelium in isolation whereas the underlying tissue also contains myriad cell types and impressive chemical and structural complexity. This review dissects the cellular and matrix components of the intestine and discusses strategies to replicate them in vitro using principles drawing from bottom-up biological self-organization and top-down bioengineering. It also covers the cellular, biochemical and biophysical features of the intestinal microenvironment and how these can be replicated in vitro by combining strategies from organoid biology with materials science. Particularly accessible chemistries that mimic the native extracellular matrix are discussed, and bioengineering approaches that aim to overcome limitations in modelling the intestine are critically evaluated. Finally, the review considers how further advances may extend the applications of intestinal models and their suitability for clinical therapies.
Collapse
Affiliation(s)
- Oliver Cameron
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
| | - Joana F. Neves
- Centre for Host‐Microbiome InteractionsKing's College LondonLondonSE1 9RTUK
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
- Department of Biomedical SciencesUniversity of LausanneLausanne1005Switzerland
| |
Collapse
|
15
|
Tearle JLE, Tang A, Vasanthakumar A, James KR. Role reversals: non-canonical roles for immune and non-immune cells in the gut. Mucosal Immunol 2024; 17:137-146. [PMID: 37967720 DOI: 10.1016/j.mucimm.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023]
Abstract
The intestine is home to an intertwined network of epithelial, immune, and neuronal cells as well as the microbiome, with implications for immunity, systemic metabolism, and behavior. While the complexity of this microenvironment has long since been acknowledged, recent technological advances have propelled our understanding to an unprecedented level. Notably, the microbiota and non-immune or structural cells have emerged as important conductors of intestinal immunity, and by contrast, cells of both the innate and adaptive immune systems have demonstrated non-canonical roles in tissue repair and metabolism. This review highlights recent works in the following two streams: non-immune cells of the intestine performing immunological functions; and traditional immune cells exhibiting non-immune functions in the gut.
Collapse
Affiliation(s)
- Jacqueline L E Tearle
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Biomedical Sciences, University of New South Wales, Australia
| | - Adelynn Tang
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| | - Ajithkumar Vasanthakumar
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia.
| | - Kylie R James
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Biomedical Sciences, University of New South Wales, Australia.
| |
Collapse
|
16
|
Yang W, Yu T, Cong Y. Stromal Cell Regulation of Intestinal Inflammatory Fibrosis. Cell Mol Gastroenterol Hepatol 2024; 17:703-711. [PMID: 38246590 PMCID: PMC10958116 DOI: 10.1016/j.jcmgh.2024.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
Intestinal inflammatory fibrosis is a severe consequence of inflammatory bowel diseases (IBDs). There is currently no cure for the treatment of intestinal fibrosis in IBD. Although inflammation is necessary for triggering fibrosis, the anti-inflammatory agents used to treat IBD are ineffective in preventing the progression of intestinal fibrosis and stricture formation once initiated, suggesting that inflammatory signals are not the sole drivers of fibrosis progression once it is established. Among multiple mechanisms involved in the initiation and progression of intestinal fibrosis in IBD, stromal cells play critical roles in mediating the process. In this review, we summarize recent progress on how stromal cells regulate intestinal fibrosis in IBD and how they are regulated by focusing on immune regulation and gut microbiota. We also outline the challenges moving forward in the field.
Collapse
Affiliation(s)
- Wenjing Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Tianming Yu
- Division of Gastroenterology and Hepatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Yingzi Cong
- Division of Gastroenterology and Hepatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Center for Human Immunobiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
17
|
Skou Hedemann M, Rønn M, Elise van der Heide M, Karlshøj Julegaard I, Olaf Nielsen M. Dietary inclusion of methanotrophic microbial cell-derived protein in the early post-weaning period sustains growth performance and intestinal health of weaner piglets. Animal 2023; 17:100798. [PMID: 37148623 DOI: 10.1016/j.animal.2023.100798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
The global demand for sustainably produced protein feeds for animal production is increasing. Methanotrophic bacteria grow on methane and convert it into microbial cell protein (MCP) that has been shown to have high nutritive value for growing pigs. The present aimed to investigate how increasing amounts of MCP in diets fed during the first 15 days after weaning affect the growth performance of piglets from weaning until day 43 postweaning. Furthermore, the effect of MCP on intestinal morphology and histopathology was assessed on day 15 after weaning. During seven consecutive weeks, approximately 480 piglets were recruited for the experiment per batch. The piglets were divided into four groups and housed in eight double pens with 60 piglets per pen. The piglets were fed one of four experimental diets with 0, 3, 6, or 10% of MCP included at the expense of fishmeal and subsequently potato protein for the first 15 days postweaning. Thereafter, all pigs were fed commercial weaner diets in two phases (days 16-30 and days 31-43) until day 43 postweaning. All diets were without medicinal zinc. Feed intake and growth were registered on double pen level during all three phases. On day 15 after weaning, 10 piglets per treatment were randomly selected, autopsied, and sampled for intestinal morphology and histopathology. Daily gain during the first 15 days postweaning tended (P = 0.09) to be affected by the inclusion of MCP in the weaning diet being lowest in the group fed 10% MCP. Treatment did not affect daily feed intake; however, Feed Conversion Ratio (FCR) was significantly affected (P = 0.003) showing the highest FCR in piglets fed 10% MCP. Growth performance was not affected by the experimental treatment during the following phases. In the small intestine, villous height tended (P = 0.09) to show a quadratic response to level of MCP in the diet with the longest villi observed after feeding 6% MCP. Dietary treatment did not affect crypt depth. The villous height to crypt depth (VC) ratio showed a quadratic response to increased dietary inclusion of MCP (P = 0.02) with piglets fed 6% MCP having the highest VC ratio. In conclusion, this study demonstrated that MCP could constitute 6% of diets as-fed (22% of total CP), at the expense of fishmeal and potato protein, for newly weaned piglets without negative effects on growth rates and FCR. The inclusion of MCP in diets for newly weaned piglets could be part of improving the sustainability of pig production.
Collapse
|
18
|
Schimpel C, Passegger C, Egger S, Tam-Amersdorfer C, Strobl H. A novel 3D cell culture model to study the human small intestinal immune landscape. Eur J Immunol 2023; 53:e2250131. [PMID: 36527196 DOI: 10.1002/eji.202250131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/21/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Several subsets of mononuclear phagocytes and DCs (MDC) populate the small intestine (SI), and these cells reportedly exert specialized functions in anti-microbial immunity and tolerance. Given the specialized phenotype of these cells, differing from other MDC family members, including their putative circulating blood precursors, local intestinal factors play key instructive roles in their differentiation. We designed an SI cell culture model composed of three intestinal epithelial cell (IEC) types, including absorptive enterocytes (E cells), antigen delivering microfold (M) cells, and mucus-producing goblet (G) cells plus T lymphocytes and soluble B cell-derived factors. This model was used to study the differentiation fate of CD34+ hematopoietic progenitor cell-derived monocyte/DC precursors. Progeny cells can be analyzed after a 3-week co-culture period, mimicking the physiologic turn-over time of intestinal MDC. A dominant monocyte differentiation pathway was suppressed, in favor of partial differentiation along DC and macrophage pathways, with low percentages of cells acquired DC or macrophage markers. Moreover, E and G cells play opposing roles in CX3CR1+ vs CD103dim cell differentiation, indicating that both together might counter-balance M/DC differentiation. Thus, SI epithelial cells suppress M/DC differentiation, supporting a key role for exogenous factors in M/DC differentiation.
Collapse
Affiliation(s)
- Christa Schimpel
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Christina Passegger
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Simone Egger
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| | - Herbert Strobl
- Medical University of Graz, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology and Pathophysiology, Graz, Austria
| |
Collapse
|
19
|
Stromal regulation of the intestinal barrier. Mucosal Immunol 2023; 16:221-231. [PMID: 36708806 DOI: 10.1016/j.mucimm.2023.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The intestinal barrier is a complex structure that allows the absorption of nutrients while ensuring protection against intestinal pathogens and balanced immunity. The development and maintenance of a functional intestinal barrier is a multifactorial process that is only partially understood. Here we review novel findings on the emerging role of mesenchymal cells in this process using insights gained from lineage tracing approaches, Cre-based gene deletion, and single-cell transcriptomics. The current evidence points toward a key organizer role for distinct mesenchymal lineages in intestinal development and homeostasis, regulating both epithelial and immune components of the intestinal barrier. We further discuss recent findings on functional mesenchymal heterogeneity and implications for intestinal regeneration and inflammatory intestinal pathologies.
Collapse
|
20
|
Bindu S, Dandapat S, Manikandan R, Dinesh M, Subbaiyan A, Mani P, Dhawan M, Tiwari R, Bilal M, Emran TB, Mitra S, Rabaan AA, Mutair AA, Alawi ZA, Alhumaid S, Dhama K. Prophylactic and therapeutic insights into trained immunity: A renewed concept of innate immune memory. Hum Vaccin Immunother 2022; 18:2040238. [PMID: 35240935 PMCID: PMC9009931 DOI: 10.1080/21645515.2022.2040238] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/18/2022] [Accepted: 02/04/2022] [Indexed: 12/16/2022] Open
Abstract
Trained immunity is a renewed concept of innate immune memory that facilitates the innate immune system to have the capacity to remember and train cells via metabolic and transcriptional events to enable them to provide nonspecific defense against the subsequent encounters with a range of pathogens and acquire a quicker and more robust immune response, but different from the adaptive immune memory. Reversing the epigenetic changes or targeting the immunological pathways may be considered potential therapeutic approaches to counteract the hyper-responsive or hypo-responsive state of trained immunity. The efficient regulation of immune homeostasis and promotion or inhibition of immune responses is required for a balanced response. Trained immunity-based vaccines can serve as potent immune stimuli and help in the clearance of pathogens in the body through multiple or heterologous effects and confer protection against nonspecific and specific pathogens. This review highlights various features of trained immunity and its applications in developing novel therapeutics and vaccines, along with certain detrimental effects, challenges as well as future perspectives.
Collapse
Affiliation(s)
- Suresh Bindu
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Satyabrata Dandapat
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Rajendran Manikandan
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Murali Dinesh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Anbazhagan Subbaiyan
- Division of Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Pashupathi Mani
- Division of Animal Biochemistry, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- Indian Council of Agricultural Research, The Trafford Group of Colleges, Manchester, UK
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, China
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangldesh
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, Australia
| | - Zainab Al Alawi
- Division of Allergy and Immunology, College of Medicine, King Faisal University, Saudi Arabia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa, Saudi Arabia
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| |
Collapse
|
21
|
Frede A, Czarnewski P, Monasterio G, Tripathi KP, Bejarano DA, Ramirez Flores RO, Sorini C, Larsson L, Luo X, Geerlings L, Novella-Rausell C, Zagami C, Kuiper R, Morales RA, Castillo F, Hunt M, Mariano LL, Hu YOO, Engblom C, Lennon-Duménil AM, Mittenzwei R, Westendorf AM, Hövelmeyer N, Lundeberg J, Saez-Rodriguez J, Schlitzer A, Das S, Villablanca EJ. B cell expansion hinders the stroma-epithelium regenerative cross talk during mucosal healing. Immunity 2022; 55:2336-2351.e12. [PMID: 36462502 DOI: 10.1016/j.immuni.2022.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 07/14/2022] [Accepted: 10/26/2022] [Indexed: 12/05/2022]
Abstract
Therapeutic promotion of intestinal regeneration holds great promise, but defining the cellular mechanisms that influence tissue regeneration remains an unmet challenge. To gain insight into the process of mucosal healing, we longitudinally examined the immune cell composition during intestinal damage and regeneration. B cells were the dominant cell type in the healing colon, and single-cell RNA sequencing (scRNA-seq) revealed expansion of an IFN-induced B cell subset during experimental mucosal healing that predominantly located in damaged areas and associated with colitis severity. B cell depletion accelerated recovery upon injury, decreased epithelial ulceration, and enhanced gene expression programs associated with tissue remodeling. scRNA-seq from the epithelial and stromal compartments combined with spatial transcriptomics and multiplex immunostaining showed that B cells decreased interactions between stromal and epithelial cells during mucosal healing. Activated B cells disrupted the epithelial-stromal cross talk required for organoid survival. Thus, B cell expansion during injury impairs epithelial-stromal cell interactions required for mucosal healing, with implications for the treatment of IBD.
Collapse
Affiliation(s)
- Annika Frede
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paulo Czarnewski
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Gustavo Monasterio
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kumar P Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David A Bejarano
- Quantitative Systems Biology, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | | | - Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ludvig Larsson
- KTH Royal Institute of Technology Stockholm, Science for Life Laboratory, Stockholm, Sweden
| | - Xinxin Luo
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laura Geerlings
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Claudio Novella-Rausell
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Chiara Zagami
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Raoul Kuiper
- Norwegian Veterinary Institute, Section for Aquatic Biosecurity Research, Elisabeth Stephansens vei 1, 1433 Ås, Norway; Core Facility for Morphologic Phenotype Analysis, Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo A Morales
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Francisca Castillo
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew Hunt
- Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | - Yue O O Hu
- Centre for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Camilla Engblom
- Department of Cell and Molecular Biology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | - Romy Mittenzwei
- Institute for Molecular Medicine and Research Center for Immunotherapy (FZI), University Medical Center Mainz, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nadine Hövelmeyer
- Institute for Molecular Medicine and Research Center for Immunotherapy (FZI), University Medical Center Mainz, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Joakim Lundeberg
- KTH Royal Institute of Technology Stockholm, Science for Life Laboratory, Stockholm, Sweden
| | - Julio Saez-Rodriguez
- Institute of Computational Biomedicine, University of Heidelberg, Heidelberg, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Srustidhar Das
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
22
|
Zhang J, Li J, Yan P, He L, Zhang X, Wang X, Shi Y, Deng L, Zhang Z, Zhao B. In-depth analysis of the relationship between bovine intestinal organoids and enteroids based on morphology and transcriptome. J Tissue Eng Regen Med 2022; 16:1032-1046. [PMID: 36128613 DOI: 10.1002/term.3351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 01/13/2023]
Abstract
Intestinal organoids and enteroids as excellent models are miniaturized and simplified for studying intestinal physiological and pathological functions, drug screening, and regenerative medicine. Recently, the application demands for organoids and enteroids in organ development and nutrition metabolism, immune and cancer research increased. But there are few comparative studies on both of them, especially in immunity and metabolism, which is also conducive to further clarifying the role of crypt stem cells and stromal cells. In our study, "natural" organoids were obtained by tissue culture from fetal bovine jejunum and enteroids were successfully isolated and cultured from organoids without supplementing exogenous factors and Matrigel. These mini-guts displayed similar features to the intestine through immunohistochemistry and transmission electron microscopy. Organoid and enteroid were systematically compared based on the transcriptome. And some of the results were verified by qRT-PCR. Our results showed KDGs (Key driver genes) (e.g., SLC13A1, HOXA7, HOXA6, HOXA5, and HOXD4) of organoids enriched in signaling pathways related to organ development and morphology and metabolism. KDGs (e.g., IL-6, PTGS2, CDH1, JUN, and EGFR) of enteroid were involved in cancer, MAPK, and immune-related signaling pathways. To the Wnt signaling pathway, highly expressed genes in organoids, including RSPO2, NOTUM, WNT6, and RSPO3, supported the homeostasis of crypt stem cells. Enteroids highly expressed CTNNB1 and WNTs. In addition, we found that organoids and enteroids carried out different functions in immunity and metabolism due to different cell compositions. Therefore, it suggested organoid is more compatible and comprehensive, and enteroid is qualified for the research of immunity and cancer.
Collapse
Affiliation(s)
- Juntao Zhang
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China.,College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Juanjuan Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Penghui Yan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Laizeng He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Xuemei Zhang
- Henan Yinfeng Biological Engineering Technology Co., LTD, Zhengzhou, Henan, China
| | - Xiaolong Wang
- Henan Yinfeng Biological Engineering Technology Co., LTD, Zhengzhou, Henan, China
| | - Yake Shi
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Lixin Deng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - ZhiPing Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A & F University, Yangling, Shaanxi, China
| |
Collapse
|
23
|
Tissue Niches Formed by Intestinal Mesenchymal Stromal Cells in Mucosal Homeostasis and Immunity. Int J Mol Sci 2022; 23:ijms23095181. [PMID: 35563571 PMCID: PMC9100044 DOI: 10.3390/ijms23095181] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 12/17/2022] Open
Abstract
The gastrointestinal tract is the largest mucosal surface in our body and accommodates the majority of the total lymphocyte population. Being continuously exposed to both harmless antigens and potentially threatening pathogens, the intestinal mucosa requires the integration of multiple signals for balancing immune responses. This integration is certainly supported by tissue-resident intestinal mesenchymal cells (IMCs), yet the molecular mechanisms whereby IMCs contribute to these events remain largely undefined. Recent studies using single-cell profiling technologies indicated a previously unappreciated heterogeneity of IMCs and provided further knowledge which will help to understand dynamic interactions between IMCs and hematopoietic cells of the intestinal mucosa. In this review, we focus on recent findings on the immunological functions of IMCs: On one hand, we discuss the steady-state interactions of IMCs with epithelial cells and hematopoietic cells. On the other hand, we summarize our current knowledge about the contribution of IMCs to the development of intestinal inflammatory conditions, such as infections, inflammatory bowel disease, and fibrosis. By providing a comprehensive list of cytokines and chemokines produced by IMCs under homeostatic and inflammatory conditions, we highlight the significant immunomodulatory and tissue niche forming capacities of IMCs.
Collapse
|
24
|
Parigi SM, Larsson L, Das S, Ramirez Flores RO, Frede A, Tripathi KP, Diaz OE, Selin K, Morales RA, Luo X, Monasterio G, Engblom C, Gagliani N, Saez-Rodriguez J, Lundeberg J, Villablanca EJ. The spatial transcriptomic landscape of the healing mouse intestine following damage. Nat Commun 2022; 13:828. [PMID: 35149721 PMCID: PMC8837647 DOI: 10.1038/s41467-022-28497-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
The intestinal barrier is composed of a complex cell network defining highly compartmentalized and specialized structures. Here, we use spatial transcriptomics to define how the transcriptomic landscape is spatially organized in the steady state and healing murine colon. At steady state conditions, we demonstrate a previously unappreciated molecular regionalization of the colon, which dramatically changes during mucosal healing. Here, we identified spatially-organized transcriptional programs defining compartmentalized mucosal healing, and regions with dominant wired pathways. Furthermore, we showed that decreased p53 activation defined areas with increased presence of proliferating epithelial stem cells. Finally, we mapped transcriptomics modules associated with human diseases demonstrating the translational potential of our dataset. Overall, we provide a publicly available resource defining principles of transcriptomic regionalization of the colon during mucosal healing and a framework to develop and progress further hypotheses.
Collapse
Affiliation(s)
- Sara M Parigi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Ludvig Larsson
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Srustidhar Das
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Ricardo O Ramirez Flores
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Annika Frede
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Kumar P Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Oscar E Diaz
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Katja Selin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Rodrigo A Morales
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Xinxin Luo
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Gustavo Monasterio
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Camilla Engblom
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Nicola Gagliani
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- I. Department of Medicine and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute and University Hospital, Stockholm, Sweden.
- Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
25
|
Shinotsuka N, Denk F. Fibroblasts: the neglected cell type in peripheral sensitisation and chronic pain? A review based on a systematic search of the literature. BMJ OPEN SCIENCE 2022; 6:e100235. [PMID: 35128075 PMCID: PMC8768938 DOI: 10.1136/bmjos-2021-100235] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/12/2021] [Indexed: 11/15/2022] Open
Abstract
Chronic pain and its underlying biological mechanisms have been studied for many decades, with a myriad of molecules, receptors and cell types known to contribute to abnormal pain sensations. Besides an obvious role for neurons, immune cells like microglia, macrophages and T cells are also important drivers of persistent pain. While neuroinflammation has therefore been widely studied in pain research, there is one cell type that appears to be rather neglected in this context: the humble fibroblast. Fibroblasts may seem unassuming but actually play a major part in regulating immune cell function and driving chronic inflammation. Here, our aim was to determine the breadth and quality of research that implicates fibroblasts in chronic pain conditions and models. OBJECTIVES We set out to analyse the current literature on this topic-using systematic screening and data extraction methods to obtain a balanced view on what has been published. METHODS We categorised the articles we included-stratifying them according to what was investigated, the estimated quality of results and any common conclusions. RESULTS We found that there has been surprisingly little research in this area: 134 articles met our inclusion criteria, only a tiny minority of which directly investigated interactions between fibroblasts and peripheral neurons. CONCLUSIONS Fibroblasts are a ubiquitous cell type and a prominent source of many proalgesic mediators in a wide variety of tissues. We think that they deserve a more central role in pain research and propose a new, testable model of how fibroblasts might drive peripheral neuron sensitisation.
Collapse
Affiliation(s)
- Naomi Shinotsuka
- Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Shizuoka, Japan
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
26
|
James KR, Elmentaite R, Teichmann SA, Hold GL. Redefining intestinal immunity with single-cell transcriptomics. Mucosal Immunol 2022; 15:531-541. [PMID: 34848830 PMCID: PMC8630196 DOI: 10.1038/s41385-021-00470-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 02/04/2023]
Abstract
The intestinal immune system represents the largest collection of immune cells in the body and is continually exposed to antigens from food and the microbiota. Here we discuss the contribution of single-cell transcriptomics in shaping our understanding of this complex system. We consider the impact on resolving early intestine development, engagement with the neighbouring microbiota, diversity of intestinal immune cells, compartmentalisation within the intestines and interactions with non-immune cells. Finally, we offer a perspective on open questions about gut immunity that evolving single-cell technologies are well placed to address.
Collapse
Affiliation(s)
- Kylie Renee James
- grid.415306.50000 0000 9983 6924Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW 2010 Australia ,grid.1005.40000 0004 4902 0432School of Medical Sciences, University of New South Wales, Sydney, NSW 2006 Australia
| | - Rasa Elmentaite
- grid.10306.340000 0004 0606 5382Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA UK
| | - Sarah Amalia Teichmann
- grid.10306.340000 0004 0606 5382Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA UK ,grid.5335.00000000121885934Theory of Condensed Matter Group, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge, NSW CB3 0HE UK
| | - Georgina Louise Hold
- grid.1005.40000 0004 4902 0432University of New South Wales Microbiome Research Centre, Sydney, NSW 2217 Australia
| |
Collapse
|
27
|
Hentschel V, Seufferlein T, Armacki M. Intestinal organoids in coculture: redefining the boundaries of gut mucosa ex vivo modeling. Am J Physiol Gastrointest Liver Physiol 2021; 321:G693-G704. [PMID: 34643092 DOI: 10.1152/ajpgi.00043.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
All-time preservation of an intact mucosal barrier is crucial to ensuring intestinal homeostasis and, hence, the organism's overall health maintenance. This complex process relies on an equilibrated signaling system between the intestinal epithelium and numerous cell populations inhabiting the gut mucosa. Any perturbations of this delicate cross talk, particularly regarding the immune cell compartment and microbiota, may sustainably debilitate the intestinal barrier function. As a final joint event, a critical rise in epithelial permeability facilitates the exposure of submucosal immunity to microbial antigens, resulting in uncontrolled inflammation, collateral tissue destruction, and dysbiosis. Organoid-derived intestinal coculture models have established themselves as convenient tools to reenact such pathophysiological events, explore interactions between selected cell populations, and assess their roles with a central focus on intestinal barrier recovery and stabilization.
Collapse
Affiliation(s)
- Viktoria Hentschel
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Milena Armacki
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
28
|
Chiaranunt P, Tai SL, Ngai L, Mortha A. Beyond Immunity: Underappreciated Functions of Intestinal Macrophages. Front Immunol 2021; 12:749708. [PMID: 34650568 PMCID: PMC8506163 DOI: 10.3389/fimmu.2021.749708] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract hosts the largest compartment of macrophages in the body, where they serve as mediators of host defense and immunity. Seeded in the complex tissue-environment of the gut, an array of both hematopoietic and non-hematopoietic cells forms their immediate neighborhood. Emerging data demonstrate that the functional diversity of intestinal macrophages reaches beyond classical immunity and includes underappreciated non-immune functions. In this review, we discuss recent advances in research on intestinal macrophage heterogeneity, with a particular focus on how non-immune functions of macrophages impact tissue homeostasis and function. We delve into the strategic localization of distinct gut macrophage populations, describe the potential factors that regulate their identity and functional heterogeneity within these locations, and provide open questions that we hope will inspire research dedicated to elucidating a holistic view on macrophage-tissue cell interactions in the body's largest mucosal organ.
Collapse
Affiliation(s)
- Pailin Chiaranunt
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Siu Ling Tai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
29
|
Hidalgo‐Garcia L, Molina‐Tijeras JA, Huertas‐Peña F, Ruiz‐Malagón AJ, Diez‐Echave P, Vezza T, Rodríguez‐Sojo MJ, Morón R, Becerra‐Massare P, Rodríguez‐Nogales A, Gálvez J, Rodríguez‐Cabezas ME, Anderson P. Intestinal mesenchymal cells regulate immune responses and promote epithelial regeneration in vitro and in dextran sulfate sodium-induced experimental colitis in mice. Acta Physiol (Oxf) 2021; 233:e13699. [PMID: 34089568 DOI: 10.1111/apha.13699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022]
Abstract
AIM Disruption of the intestinal mucosal tolerance, that is, the immunological unresponsiveness to innocuous food antigens and the commensal microbiota, in the colon is associated with several chronic diseases including inflammatory bowel disease (IBD). Understanding the mechanisms responsible for intestinal mucosal tolerance has potential translational value for its therapy and management. Human intestinal mesenchymal cells (iMCs) play important roles in colonic mucosal tolerance, but further studies on their tissue regenerative and immunomodulatory capacities are necessary in order to fully understand their function in health and disease. METHODS In this study, we have isolated and analysed the capacity of human iMCs to promote wound healing and modulate immune responses in vitro and in vivo, using the dextran sulfate sodium (DSS)-induced colitis model. RESULTS Cultured iMCs were CD45- CD73+ CD90+ CD105+ and accelerated the wound closure in a normal colon mucosa (NCM) 356 human epithelial cell wound healing assay. Furthermore, iMCs blocked the LPS-mediated induction of TNF-α in THP-1 macrophages and inhibited the proliferation of peripheral blood mononuclear cells, partly through the induction of indoleamine-2,3-dioxygenase. In DSS colitic mice, iMCs administration reduced the disease activity index and ameliorated intestinal tissue damage and permeability. Furthermore, iMCs reduced intestinal inflammation, evidenced by a decreased mRNA expression of pro-inflammatory cytokines, reduced IL-1β secretion by intestinal explants and inhibited colonic iNOS protein expression. CONCLUSIONS Our data show that human iMCs isolated from the noninflamed intestine possess tissue-regenerative and immunomodulatory capacities that could potentially be harnessed/restored in order to reduce IBD severity.
Collapse
Affiliation(s)
- Laura Hidalgo‐Garcia
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - José Alberto Molina‐Tijeras
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Francisco Huertas‐Peña
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio de Cirugía Hospital Universitario Virgen de las Nieves Granada Spain
| | - Antonio Jesús Ruiz‐Malagón
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Patricia Diez‐Echave
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Teresa Vezza
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - María Jesús Rodríguez‐Sojo
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Rocío Morón
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio Farmacia Hospitalaria Hospital Universitario Clínico San Cecilio Granada Spain
| | | | - Alba Rodríguez‐Nogales
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio de Digestivo Hospital Universitario Virgen de las Nieves Granada Spain
| | - Julio Gálvez
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBER‐EHD) University of Granada Granada Spain
| | - María Elena Rodríguez‐Cabezas
- Department of Pharmacology Center for Biomedical Research (CIBM) University of Granada Granada Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
| | - Per Anderson
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA) Granada Spain
- Servicio de Análisis Clínicos e Inmunología Hospital Universitario Virgen de las Nieves Granada Spain
| |
Collapse
|
30
|
Jacobse J, Li J, Rings EHHM, Samsom JN, Goettel JA. Intestinal Regulatory T Cells as Specialized Tissue-Restricted Immune Cells in Intestinal Immune Homeostasis and Disease. Front Immunol 2021; 12:716499. [PMID: 34421921 PMCID: PMC8371910 DOI: 10.3389/fimmu.2021.716499] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/16/2021] [Indexed: 12/28/2022] Open
Abstract
FOXP3+ regulatory T cells (Treg cells) are a specialized population of CD4+ T cells that restrict immune activation and are essential to prevent systemic autoimmunity. In the intestine, the major function of Treg cells is to regulate inflammation as shown by a wide array of mechanistic studies in mice. While Treg cells originating from the thymus can home to the intestine, the majority of Treg cells residing in the intestine are induced from FOXP3neg conventional CD4+ T cells to elicit tolerogenic responses to microbiota and food antigens. This process largely takes place in the gut draining lymph nodes via interaction with antigen-presenting cells that convert circulating naïve T cells into Treg cells. Notably, dysregulation of Treg cells leads to a number of chronic inflammatory disorders, including inflammatory bowel disease. Thus, understanding intestinal Treg cell biology in settings of inflammation and homeostasis has the potential to improve therapeutic options for patients with inflammatory bowel disease. Here, the induction, maintenance, trafficking, and function of intestinal Treg cells is reviewed in the context of intestinal inflammation and inflammatory bowel disease. In this review we propose intestinal Treg cells do not compose fixed Treg cell subsets, but rather (like T helper cells), are plastic and can adopt different programs depending on microenvironmental cues.
Collapse
Affiliation(s)
- Justin Jacobse
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
| | - Edmond H. H. M. Rings
- Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
- Department of Pediatrics, Sophia Children’s Hospital, Erasmus University, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Janneke N. Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeremy A. Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
31
|
MiR-210-5p regulates STAT3 activation by targeting STAT5A in the differentiation of dermal fibroblasts. 3 Biotech 2021; 11:243. [PMID: 33968586 DOI: 10.1007/s13205-021-02777-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 04/05/2021] [Indexed: 10/21/2022] Open
Abstract
Elucidating the molecular mechanism of the microRNAs in skin fibrosis is critical for identifying a novel therapeutic strategy for hypertrophic scar (HS). In this study, it was shown that miR-210-5p is induced by TGFβ, and that overexpression of miR-210-5p promoted the differentiation of human dermal fibroblasts (HDFs) into myofibroblasts. STAT5A is required for TGFβ-induced STAT3 activity. Here, we show that miR-210-5p attenuated TGFβ-induced STAT3 signaling pathway by suppressing the expression of STAT5A. Taken together, the present study suggests that TGFβ-induced miR-210-5p reduced STAT5A expression, leading to aberrant activation of STAT3, and facilitate skin fibrosis in HDFs.
Collapse
|
32
|
Malijauskaite S, Connolly S, Newport D, McGourty K. Gradients in the in vivo intestinal stem cell compartment and their in vitro recapitulation in mimetic platforms. Cytokine Growth Factor Rev 2021; 60:76-88. [PMID: 33858768 DOI: 10.1016/j.cytogfr.2021.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Intestinal tissue, and specifically its mucosal layer, is a complex and gradient-rich environment. Gradients of soluble factor (BMP, Noggin, Notch, Hedgehog, and Wnt), insoluble extracellular matrix proteins (laminins, collagens, fibronectin, and their cognate receptors), stromal stiffness, oxygenation, and sheer stress induced by luminal fluid flow at the crypt-villus axis controls and supports healthy intestinal tissue homeostasis. However, due to current technological challenges, very few of these features have so far been included in in vitro intestinal tissue mimetic platforms. In this review, the tightly defined and dynamic microenvironment of the intestinal tissue is presented in detail. Additionally, the authors introduce the current state-of-the-art intestinal tissue mimetic platforms, as well as the design drawbacks and challenges they face while attempting to capture the complexity of the intestinal tissue's physiology. Finally, the compositions of an "idealized" mimetic system is presented to guide future developmental efforts.
Collapse
Affiliation(s)
- Sigita Malijauskaite
- Dept. of Chemical Sciences, University of Limerick, Limerick, Ireland; Bernal Institute, University of Limerick, Limerick, Ireland.
| | - Sinead Connolly
- Bernal Institute, University of Limerick, Limerick, Ireland; School of Engineering, University of Limerick, Limerick, Ireland.
| | - David Newport
- Bernal Institute, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland; School of Engineering, University of Limerick, Limerick, Ireland.
| | - Kieran McGourty
- Dept. of Chemical Sciences, University of Limerick, Limerick, Ireland; Bernal Institute, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland.
| |
Collapse
|
33
|
Bigaeva E, Uniken Venema WTC, Weersma RK, Festen EAM. Understanding human gut diseases at single-cell resolution. Hum Mol Genet 2020; 29:R51-R58. [PMID: 32588873 PMCID: PMC7530522 DOI: 10.1093/hmg/ddaa130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
Our understanding of gut functioning and pathophysiology has grown considerably in the past decades, and advancing technologies enable us to deepen this understanding. Single-cell RNA sequencing (scRNA-seq) has opened a new realm of cellular diversity and transcriptional variation in the human gut at a high, single-cell resolution. ScRNA-seq has pushed the science of the digestive system forward by characterizing the function of distinct cell types within complex intestinal cellular environments, by illuminating the heterogeneity within specific cell populations and by identifying novel cell types in the human gut that could contribute to a variety of intestinal diseases. In this review, we highlight recent discoveries made with scRNA-seq that significantly advance our understanding of the human gut both in health and across the spectrum of gut diseases, including inflammatory bowel disease, colorectal carcinoma and celiac disease.
Collapse
Affiliation(s)
- Emilia Bigaeva
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Werna T C Uniken Venema
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ Groningen, The Netherlands
| |
Collapse
|
34
|
Distinct fibroblast subsets regulate lacteal integrity through YAP/TAZ-induced VEGF-C in intestinal villi. Nat Commun 2020; 11:4102. [PMID: 32796823 PMCID: PMC7428020 DOI: 10.1038/s41467-020-17886-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/23/2020] [Indexed: 02/08/2023] Open
Abstract
Emerging evidence suggests that intestinal stromal cells (IntSCs) play essential roles in maintaining intestinal homeostasis. However, the extent of heterogeneity within the villi stromal compartment and how IntSCs regulate the structure and function of specialized intestinal lymphatic capillary called lacteal remain elusive. Here we show that selective hyperactivation or depletion of YAP/TAZ in PDGFRβ+ IntSCs leads to lacteal sprouting or regression with junctional disintegration and impaired dietary fat uptake. Indeed, mechanical or osmotic stress regulates IntSC secretion of VEGF-C mediated by YAP/TAZ. Single-cell RNA sequencing delineated novel subtypes of villi fibroblasts that upregulate Vegfc upon YAP/TAZ activation. These populations of fibroblasts were distributed in proximity to lacteal, suggesting that they constitute a peri-lacteal microenvironment. Our findings demonstrate the heterogeneity of IntSCs and reveal that distinct subsets of villi fibroblasts regulate lacteal integrity through YAP/TAZ-induced VEGF-C secretion, providing new insights into the dynamic regulatory mechanisms behind lymphangiogenesis and lymphatic remodeling. Intestinal stromal cells (IntSCs) play essential roles in maintaining intestinal homeostasis. Here the authors show that VEGF-C expression in specialized IntSCs is regulated by YAP/TAZ, and VEGF-C is responsible for maintaining lacteal integrity, thus influencing dietary fat drainage into lacteals.
Collapse
|
35
|
Coakley G, Harris NL. The Intestinal Epithelium at the Forefront of Host-Helminth Interactions. Trends Parasitol 2020; 36:761-772. [PMID: 32713764 DOI: 10.1016/j.pt.2020.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Gastrointestinal helminth infection still constitutes a major public health issue, particularly in the developing world. As these parasites can undergo a large part of their lifecycle within the intestinal tract the host has developed various structural and cellular specializations at the epithelial barrier to contend with infection. Detailed characterization of these cells will provide important insights about their contributions to the protective responses mediated against helminths. Here, we discuss how key components of the intestinal epithelium may function to limit the initial establishment of helminths, and how these cells are altered during an active response to infection.
Collapse
Affiliation(s)
- Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, Victoria, Australia.
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Sato Y, Joumura T, Takekuma Y, Sugawara M. Transfer of orally administered hyaluronan to the lymph. Eur J Pharm Biopharm 2020; 154:210-213. [PMID: 32681965 DOI: 10.1016/j.ejpb.2020.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/16/2020] [Accepted: 07/07/2020] [Indexed: 11/28/2022]
Abstract
Hyaluronan (HA) has been widely used in medicines, cosmetics and supplements for health and beauty maintenance. Oral administration is the most desirable and convenient means for consumers. The intestine plays an important role in immune system. We hypothesized that orally administered HA would be transferred to both blood and lymph. In this study, we investigated how orally administered HA was absorbed from the gastrointestinal tract and how much HA was incorporated. Four HA formulations, HA-2,000, 8,000, 50,000 and 300,000, were administered to rats, and concentrations of HA in blood and lymph were determined. In the HA-2,000 group, the HA plasma concentration increased after oral administration. The highest lymph concentration of HA was also obtained by administration of HA-2,000. The plasma and lymph concentrations slightly increased after oral administration in the HA-8,000 group. On the other hand, little absorption was found in the HA-50,000 and 300,000 groups. It is speculated that smaller molecules of HA are more easily absorbed. HA-2,000 was absorbed mainly through the portal vein and through the lymph in gastrointestinal absorption. This is the first report showing that HAs, large molecular weight and water-soluble molecules, after oral administration are transferred not only into blood but also into lymph.
Collapse
Affiliation(s)
- Yuki Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Tatsuru Joumura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Yoh Takekuma
- Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-0848, Japan
| | - Mitsuru Sugawara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan; Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-0848, Japan.
| |
Collapse
|
37
|
Ferber S, Gonzalez RJ, Cryer AM, von Andrian UH, Artzi N. Immunology-Guided Biomaterial Design for Mucosal Cancer Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903847. [PMID: 31833592 DOI: 10.1002/adma.201903847] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/11/2019] [Indexed: 05/23/2023]
Abstract
Cancer of mucosal tissues is a major cause of worldwide mortality for which only palliative treatments are available for patients with late-stage disease. Engineered cancer vaccines offer a promising approach for inducing antitumor immunity. The route of vaccination plays a major role in dictating the migratory pattern of lymphocytes, and thus vaccine efficacy in mucosal tissues. Parenteral immunization, specifically subcutaneous and intramuscular, is the most common vaccination route. However, this induces marginal mucosal protection in the absence of tissue-specific imprinting signals. To circumvent this, the mucosal route can be utilized, however degradative mucosal barriers must be overcome. Hence, vaccine administration route and selection of materials able to surmount transport barriers are important considerations in mucosal cancer vaccine design. Here, an overview of mucosal immunity in the context of cancer and mucosal cancer clinical trials is provided. Key considerations are described regarding the design of biomaterial-based vaccines that will afford antitumor immune protection at mucosal surfaces, despite limited knowledge surrounding mucosal vaccination, particularly aided by biomaterials and mechanistic immune-material interactions. Finally, an outlook is given of how future biomaterial-based mucosal cancer vaccines will be shaped by new discoveries in mucosal vaccinology, tumor immunology, immuno-therapeutic screens, and material-immune system interplay.
Collapse
Affiliation(s)
- Shiran Ferber
- Department of Medicine, Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rodrigo J Gonzalez
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexander M Cryer
- Department of Medicine, Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Boston, MA, 02139, USA
| | - Natalie Artzi
- Department of Medicine, Engineering in Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02139, USA
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Zhang J, Penny J, Lu JR. Development of a novel in vitro 3D intestinal model for permeability evaluations. Int J Food Sci Nutr 2019; 71:549-562. [DOI: 10.1080/09637486.2019.1700940] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Jing Zhang
- Biological Physics Laboratory, School of Physics and Astronomy, Faculty of Sciences and Engineering, University of Manchester, Manchester, UK
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jian R. Lu
- Biological Physics Laboratory, School of Physics and Astronomy, Faculty of Sciences and Engineering, University of Manchester, Manchester, UK
| |
Collapse
|
39
|
Zhu F, Fan H, Liu XX. Role of miR-155 in pathogenesis of inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2019; 27:1070-1075. [DOI: 10.11569/wcjd.v27.i17.1070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a non-infectious, non-specific intestinal inflammatory disease, and its incidence is increasing year by year. A better understanding of the pathogenesis of IBD can help to find more suitable treatment methods for this disease. Recently, a large number of studies have shown that miR-155 plays an important role in the pathogenesis of IBD. It can regulate TH17 differentiation through the Jarid2/notch1 signaling pathway and by regulating type 2 macrophage differentiation. It also regulates T-regulatory cells by inhibiting cytotoxic T lymphocyte-associated antigen-4; intestinal myelofibrosis through suppressor of cytokine signaling 1; and DNA double-strand break deposition to affect intestinal inflammation. This article reviews the immune mechanism of action of miR-155 in IBD.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xing-Xing Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
40
|
Ren C, Dokter‐Fokkens J, Figueroa Lozano S, Zhang Q, de Haan BJ, Zhang H, Faas MM, de Vos P. Fibroblasts Impact Goblet Cell Responses to Lactic Acid Bacteria After Exposure to Inflammatory Cytokines and Mucus Disruptors. Mol Nutr Food Res 2019; 63:e1801427. [PMID: 30977971 PMCID: PMC6618108 DOI: 10.1002/mnfr.201801427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/21/2019] [Indexed: 12/15/2022]
Abstract
SCOPE Mucus produced by goblet cells contributes to gut barrier function. Lactic acid bacteria (LAB) have been shown to impact mucus production. It is not completely known whether mucus production is influenced by the abundantly present fibroblasts in the intestine. METHODS AND RESULTS The influence of fibroblasts on mucus-related genes including mucin-2 (MUC2), trefoil factor 3 (TFF3), resistin-like molecule β (RETNLB), carbohydrate sulfotransferase 5 (CHST5), and galactose-3-O-sulfotransferase 2 (GAL3ST2) is examined after co-culture of LS174T-goblet cells and CCD-18Co colonic fibroblasts in the presence and absence of LAB-strains known to impact mucus function. This is also tested after exposure to TNF-α, IL-13, or the mucin synthesis inhibitor tunicamycin (Tm). Effects of fibroblasts are treatment duration- and bacterial species-dependent under homeostatic conditions. During TNF-α challenge, fibroblasts reverse Lactobacillus (L.) rhamnosus CCFM237-elicited declined TFF3 expression. After IL-13 exposure, L. rhamnosus CCFM237 and L. fermentum CCFM787 attenuate enhanced TFF3 and RETNLB expression, respectively, only in the presence of fibroblasts. LAB has no effects on Tm-induced decreased expression of goblet cell-related genes regardless of the presence of fibroblasts. CONCLUSION It is demonstrated that goblet cell-fibroblast crosstalk impacts mucus synthesis and influences the effects of LAB on goblet cell-related genes. Effects are LAB-species and stressor dependent.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
- School of Food Science and TechnologyJiangnan UniversityWuxi214122China
| | - Jelleke Dokter‐Fokkens
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Susana Figueroa Lozano
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Qiuxiang Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxi214122China
| | - Bart J. de Haan
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Hao Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxi214122China
| | - Marijke M. Faas
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center Groningen9700RBGroningenThe Netherlands
| |
Collapse
|
41
|
Walsh KT, Zemper AE. The Enteric Nervous System for Epithelial Researchers: Basic Anatomy, Techniques, and Interactions With the Epithelium. Cell Mol Gastroenterol Hepatol 2019; 8:369-378. [PMID: 31108231 PMCID: PMC6718943 DOI: 10.1016/j.jcmgh.2019.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 02/08/2023]
Abstract
The intestinal epithelium does not function in isolation, but interacts with many components including the Enteric Nervous System (ENS). Understanding ENS and intestinal epithelium interactions requires multidisciplinary approaches to uncover cells involved, mechanisms used, and the ultimate influence on intestinal physiology. This review is intended to serve as a reference for epithelial biologists interested in studying these interactions. With this in mind, this review aims to summarize the basic anatomy of the epithelium and ENS, mechanisms by which they interact, and techniques used to study these interactions. We highlight in vitro, ex vivo and in vivo techniques. Additionally, ENS influence on epithelial proliferation and gene expression within stem and differentiated cells as well as gastrointestinal cancer are discussed.
Collapse
Key Words
- 5-ht, 5-hydroxytryptamine
- 5-ht3r, 5-hydroxytryptamine 3 receptor
- ach, acetylcholine
- aitc, allyl isothicyanate
- cpi, crypt proliferation index
- eec, enteroendocrine cell
- ens, enteric nervous system
- gi, gastrointestinal
- hio, human intestinal organoid
- isc, intestinal stem cell
- lgr5, leucine-rich repeat–containing g protein–coupled receptor
- ne, norepinephrine
- ngf, nerve growth factor
- si, small intestine
- ta, transit-amplifying
Collapse
Affiliation(s)
- Kathleen T. Walsh
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon,Institute of Neuroscience, University of Oregon, Eugene, Oregon,Department of Biology, University of Oregon, Eugene, Oregon
| | - Anne E. Zemper
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon,Department of Biology, University of Oregon, Eugene, Oregon,Correspondence Address correspondence to: Anne E. Zemper, PhD, University of Oregon, 218 Streisinger Hall, 1370 Franklin Boulevard, Eugene, Oregon 97401. fax: (541) 346–6056.
| |
Collapse
|
42
|
West NR. Coordination of Immune-Stroma Crosstalk by IL-6 Family Cytokines. Front Immunol 2019; 10:1093. [PMID: 31156640 PMCID: PMC6529849 DOI: 10.3389/fimmu.2019.01093] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022] Open
Abstract
Stromal cells are a subject of rapidly growing immunological interest based on their ability to influence virtually all aspects of innate and adaptive immunity. Present in every bodily tissue, stromal cells complement the functions of classical immune cells by sensing pathogens and tissue damage, coordinating leukocyte recruitment and function, and promoting immune response resolution and tissue repair. These diverse roles come with a price: like classical immune cells, inappropriate stromal cell behavior can lead to various forms of pathology, including inflammatory disease, tissue fibrosis, and cancer. An important immunological function of stromal cells is to act as information relays, responding to leukocyte-derived signals and instructing leukocyte behavior in kind. In this regard, several members of the interleukin-6 (IL-6) cytokine family, including IL-6, IL-11, oncostatin M (OSM), and leukemia inhibitory factor (LIF), have gained recognition as factors that mediate crosstalk between stromal and immune cells, with diverse roles in numerous inflammatory and homeostatic processes. This review summarizes our current understanding of how IL-6 family cytokines control stromal-immune crosstalk in health and disease, and how these interactions can be leveraged for clinical benefit.
Collapse
Affiliation(s)
- Nathaniel R West
- Department of Cancer Immunology, Genentech, South San Francisco, CA, United States
| |
Collapse
|
43
|
Misawa MYO, Silvério Ruiz KG, Nociti FH, Albiero ML, Saito MT, Nóbrega Stipp R, Condino-Neto A, Holzhausen M, Palombo H, Villar CC. Periodontal ligament-derived mesenchymal stem cells modulate neutrophil responses via paracrine mechanisms. J Periodontol 2019; 90:747-755. [PMID: 30644104 DOI: 10.1002/jper.18-0220] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 11/03/2018] [Accepted: 11/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mesenchymal stem cells differentiate into distinct mesenchymal cell lineages and regulate the immune response. The aim of this study was to determine whether periodontal ligament-derived mesenchymal stem cells (PDLSCs) have the ability to modulate neutrophil responses via paracrine mechanisms. METHODS CD105-enriched PDLSCs were seeded for 24 h and challenged with Porphyromonas gingivalis total protein extract (PgPE) (0 or 2 ug/mL) for 3 h. Cells were then washed and further cultured for 18 h and the supernatants were collected and stored. Next, neutrophil-differentiated human promyelocytic leukemia HL-60 cells (HL60D) were treated with PDLSCs supernatants and HL-60D activation and functional responses were determined. RESULTS PgPE treatment induced higher secretion of inflammatory markers and chemokines by PDLSCs, including RANTES, eotaxin, interferon (IFN)-γ- inducible protein 10 (IP-10), monocyte chemoattractant protein-1 (MCP-1), IFN-γ, interleukin (IL)-6, IL-8 and IL-1ra (P < 0.05). HL-60D recruitment rate was increased by 4.7 ± 1.09-fold when exposed to PgPE-treated PDLSCs supernatants. PgPE-treated PDLSCs supernatants promoted a 1.78 ± 1.04-fold increase in the production of intracellular reactive oxygen species (ROS) by PMA-stimulated HL-60D, whereas PgPE-untreated PDLSCs supernatants led to a 16% reduction in intracellular ROS. In sharp contrast, neither PgPE-untreated nor PgPE-treated PDLSCs supernatants altered tumor necrosis factor (TNF)-α and IL-1β secretion by HL-60D cells. CONCLUSION Together, these findings suggest an important role of PDLSCs in the recognition of P. gingivalis, paracrine recruitment and activation of antimicrobial mechanisms in innate immune cells, without interfering in cytokine responses.
Collapse
Affiliation(s)
- Mônica Yuri Orita Misawa
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Karina Gonzales Silvério Ruiz
- Division of Periodontics, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Francisco Humberto Nociti
- Division of Periodontics, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Mayra Laino Albiero
- Division of Periodontics, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Miki Taketomi Saito
- Division of Periodontics, Department of Prosthodontics and Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Rafael Nóbrega Stipp
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Antônio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marinella Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Henrique Palombo
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Cristina Cunha Villar
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil.,Department of Periodontics, UTHSC, San Antonio Dental School, San Antonio, TX, USA
| |
Collapse
|
44
|
Hamada A, Torre C, Drancourt M, Ghigo E. Trained Immunity Carried by Non-immune Cells. Front Microbiol 2019; 9:3225. [PMID: 30692968 PMCID: PMC6340064 DOI: 10.3389/fmicb.2018.03225] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
“Trained immunity” is a term proposed by Netea to describe the ability of an organism to develop an exacerbated immunological response to protect against a second infection independent of the adaptative immunity. This immunological memory can last from 1 week to several months and is only described in innate immune cells such as monocytes, macrophages, and natural killer cells. Paradoxically, the lifespan of these cells in the blood is shorter than the duration of trained immunity. This observation suggested that trained immunity could be carried by long lifespan cells such as stem cells and non-immune cells like fibroblasts. It is now evident that in addition to performing their putative function in the development and maintenance of tissue homeostasis, non-immune cells also play an important role in the response to pathogens by producing anti-microbial factors, with long-term inflammation suggesting that non-immune cells can be trained to confer long-lasting immunological memory. This review provides a summary of the current relevant knowledge about the cells which possess immunological memory and discusses the possibility that non-immune cells may carry immunological memory and mechanisms that might be involved.
Collapse
Affiliation(s)
- Attoumani Hamada
- IRD, MEPHI, Institut Hospitalier Universitaire Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Cédric Torre
- IRD, MEPHI, Institut Hospitalier Universitaire Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Michel Drancourt
- IRD, MEPHI, Institut Hospitalier Universitaire Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Eric Ghigo
- IRD, MEPHI, Institut Hospitalier Universitaire Méditerranée Infection, Aix-Marseille University, Marseille, France
| |
Collapse
|
45
|
Cen SY, Moreau JM, Furlonger C, Berger A, Paige CJ. Differential regulation of IgA + B cells in vitro by stromal cells from distinctive anatomical compartments. J Leukoc Biol 2018; 105:507-518. [PMID: 30576006 DOI: 10.1002/jlb.1a0517-172rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/06/2018] [Accepted: 11/20/2018] [Indexed: 12/19/2022] Open
Abstract
B cell development is regulated by stromal cells (SCs) that form a supportive microenvironment. These SCs along with other cell types produce cytokines, chemokines, and adhesion molecules that guide B cell commitment and differentiation. BM, spleen (Sp), and the gut lamina propria (LP) constitute distinctive anatomical compartments that support B cell differentiation. In order to characterize and compare the signals necessary to generate IgA+ B cells, we developed an in vitro system to co-culture gut LP, BM, or Sp-derived SCs with B lineage cells. Using this co-culture system, we found that gut LP SCs promote IgA+ B cell accumulation through the production of soluble stimulatory factors. In contrast to gut LP SCs, BM and splenic SCs were found to impair IgA+ B cell accumulation in vitro. Taken together, these observations provide new insights into how SCs derived from different anatomical locations shape IgA+ B cell responses.
Collapse
Affiliation(s)
- Selena Y Cen
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Joshua M Moreau
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Caren Furlonger
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alexandra Berger
- Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Christopher J Paige
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Ontario Cancer Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Christodoulou-Vafeiadou E, Ioakeimidis F, Andreadou M, Giagkas G, Stamatakis G, Reczko M, Samiotaki M, Papanastasiou AD, Karakasiliotis I, Kontoyiannis DL. Divergent Innate and Epithelial Functions of the RNA-Binding Protein HuR in Intestinal Inflammation. Front Immunol 2018; 9:2732. [PMID: 30532756 PMCID: PMC6265365 DOI: 10.3389/fimmu.2018.02732] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/06/2018] [Indexed: 12/17/2022] Open
Abstract
HuR is an abundant RNA-binding protein acting as a post-transcriptional regulator of many RNAs including mRNAs encoding inflammatory mediators, cytokines, death signalers and cell cycle regulators. In the context of intestinal pathologies, elevated HuR is considered to enhance the stability and the translation of pro-tumorigenic mRNAs providing the rationale for its pharmacological targeting. However, HuR also possesses specific regulatory functions for innate immunity and cytokine mRNA control which can oppose intestinal inflammation and tumor promotion. Here, we aim to identify contexts of intestinal inflammation where the innate immune and the epithelial functions of HuR converge or diverge. To address this, we use a disease-oriented phenotypic approach using mice lacking HuR either in intestinal epithelia or myeloid-derived immune compartments. These mice were compared for their responses to (a) Chemically induced Colitis; (b) Colitis- associated Cancer (CAC); (c) T-cell mediated enterotoxicity; (d) Citrobacter rodentium-induced colitis; and (e) TNF-driven inflammatory bowel disease. Convergent functions of epithelial and myeloid HuR included their requirement for suppressing inflammation in chemically induced colitis and their redundancies in chronic TNF-driven IBD and microbiota control. In the other contexts however, their functions diversified. Epithelial HuR was required to protect the epithelial barrier from acute inflammatory or infectious degeneration but also to promote tumor growth. In contrast, myeloid HuR was required to suppress the beneficial inflammation for pathogen clearance and tumor suppression. This cellular dichotomy in HuR's functions was validated further in mice engineered to express ubiquitously higher levels of HuR which displayed diminished pathologic and beneficial inflammatory responses, resistance to epithelial damage yet a heightened susceptibility to CAC. Our study demonstrates that epithelial and myeloid HuR affect different cellular dynamics in the intestine that need to be carefully considered for its pharmacological exploitation and points toward potential windows for harnessing HuR functions in intestinal inflammation.
Collapse
Affiliation(s)
| | - Fotis Ioakeimidis
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Margarita Andreadou
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Giorgos Giagkas
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - George Stamatakis
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Martin Reczko
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Martina Samiotaki
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | | | - Ioannis Karakasiliotis
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Dimitris L Kontoyiannis
- Division of Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece.,Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
47
|
Patel MV, Shen Z, Rossoll RM, Wira CR. Estradiol-regulated innate antiviral responses of human endometrial stromal fibroblasts. Am J Reprod Immunol 2018; 80:e13042. [PMID: 30295964 PMCID: PMC6275105 DOI: 10.1111/aji.13042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022] Open
Abstract
PROBLEM The contribution of fibroblasts to innate immune protection of the human female reproductive tract (FRT) against viral pathogens is relatively unknown. METHOD OF STUDY Endometrial (EM), endocervical (Cx) and ectocervical (ECx) fibroblasts were isolated from hysterectomy patients and grown in vitro. Fibroblasts were treated with the viral mimic poly (I:C) in the presence or absence of the sex hormone estradiol (E2 ), with gene expression measured by real-time RT-PCR and protein secretion by ELISA. RESULTS Poly (I:C) induced the expression of the interferon-stimulated genes (ISG) MxA, OAS2 and APOBEC3G, and the cytokines MCP-1, IL-8, IL-6, CCL20, IFNβ and RANTES by fibroblasts from all three sites. ISG upregulation was dependent upon Type I IFN signaling. E2 inhibited the poly (I:C)-induced upregulation of MxA and OAS2 in EM fibroblasts, but not Cx or ECx fibroblasts. E2 upregulated SDF-1α by EM fibroblasts but had no effect on secretion of other cytokines either alone or in the presence of poly (I:C). Conditioned media (CM) from poly (I:C)-treated or E2 -treated fibroblasts significantly reduced HIV infection of CD4+ T cells. CONCLUSION Stromal fibroblasts represent a level of innate immune protection against viral pathogens in the FRT beyond that seen with epithelial cells and immune cells. Our findings indicate that fibroblasts FRT are selectively responsive to E2 , capable of initiating an antiviral response against viral pathogens and may play a role in preventing HIV infection of CD4+ T cells.
Collapse
Affiliation(s)
- Mickey V. Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Zheng Shen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Richard M. Rossoll
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Charles R. Wira
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| |
Collapse
|
48
|
Hodge R, Mandle HB, Ray S, Tandon S, Peterson M, Henry A, Jahan FA, Bostick RM, Baron JA, Barry EL, Yacoub R, Rutherford RE, Seabrook ME, Fedirko V. Effects of Supplemental Calcium and Vitamin D on Expression of Toll-Like Receptors and Phospho-IKKα/β in the Normal Rectal Mucosa of Colorectal Adenoma Patients. Cancer Prev Res (Phila) 2018; 11:707-716. [PMID: 30209117 PMCID: PMC6214739 DOI: 10.1158/1940-6207.capr-18-0123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/19/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Chronic inflammation in the colorectum, a significant contributor to colorectal carcinogenesis, can be triggered by the activation of proinflammatory signaling pathways such as those initiated by Toll-like receptors (TLR) and nuclear factor κB (NF-κB). Although experimental evidence supports calcium and vitamin D potentially modifying these proinflammatory pathways in the colorectum, human data in these regards are scarce. We investigated supplemental calcium (1,200 mg daily) and/or vitamin D3 (1,000 IU daily) effects on inflammatory signaling pathway-related biomarkers in a subset of 105 participants from a colorectal adenoma recurrence chemoprevention clinical trial. We assessed expression of TLR4 and TLR5, which recognize the bacterial components lipopolysaccharides and flagellin, respectively, and phospho-IKKα/β (pIKKα/β), a biomarker of inflammation, in the normal-appearing rectal crypt epithelium and stroma using standardized, automated immunohistochemistry and quantitative image analysis. Following 1 year of treatment, TLR4, TLR5, and pIKKα/β expression in the rectal mucosa did not statistically significantly change with vitamin D or calcium supplementation, taken alone or in combination. Several baseline participant characteristics, including body mass index, history of sessile serrated adenomas, high red/processed meat intake, and high levels of rectal epithelial cell proliferation (as measured by MIB-1/Ki-67), were associated with higher baseline expression of TLRs or pIKKα/β. Our findings suggest that vitamin D and calcium may have no substantial effect on the investigated biomarkers. However, several modifiable lifestyle factors may be associated with TLRs and pIKKα/β expression in the normal rectal mucosa, supporting their future investigation as potentially treatable, preneoplastic risk factors for colorectal neoplasms. Cancer Prev Res; 11(11); 707-16. ©2018 AACR.
Collapse
Affiliation(s)
- Rebecca Hodge
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Hannah B Mandle
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Stephen Ray
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Sonia Tandon
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Meaghan Peterson
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Abigail Henry
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ferdous A Jahan
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Roberd M Bostick
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - John A Baron
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Elizabeth L Barry
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Rami Yacoub
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Robin E Rutherford
- Division of Digestive Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | | | - Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
49
|
Disson O, Blériot C, Jacob JM, Serafini N, Dulauroy S, Jouvion G, Fevre C, Gessain G, Thouvenot P, Eberl G, Di Santo JP, Peduto L, Lecuit M. Peyer's patch myeloid cells infection by Listeria signals through gp38 + stromal cells and locks intestinal villus invasion. J Exp Med 2018; 215:2936-2954. [PMID: 30355616 PMCID: PMC6219733 DOI: 10.1084/jem.20181210] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/05/2018] [Accepted: 09/28/2018] [Indexed: 12/12/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes (Lm) crosses the intestinal villus epithelium via goblet cells (GCs) upon the interaction of Lm surface protein InlA with its receptor E-cadherin. Here, we show that Lm infection accelerates intestinal villus epithelium renewal while decreasing the number of GCs expressing luminally accessible E-cadherin, thereby locking Lm portal of entry. This novel innate immune response to an enteropathogen is triggered by the infection of Peyer's patch CX3CR1+ cells and the ensuing production of IL-23. It requires STAT3 phosphorylation in epithelial cells in response to IL-22 and IL-11 expressed by lamina propria gp38+ stromal cells. Lm-induced IFN-γ signaling and STAT1 phosphorylation in epithelial cells is also critical for Lm-associated intestinal epithelium response. GC depletion also leads to a decrease in colon mucus barrier thickness, thereby increasing host susceptibility to colitis. This study unveils a novel innate immune response to an enteropathogen, which implicates gp38+ stromal cells and locks intestinal villus invasion, but favors colitis.
Collapse
Affiliation(s)
- Olivier Disson
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Camille Blériot
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Jean-Marie Jacob
- Institut Pasteur, Stroma, Inflammation and Tissue Repair Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1224, Paris, France
| | - Nicolas Serafini
- Institut Pasteur, Innate Immunity Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1223, Paris, France
| | - Sophie Dulauroy
- Institut National de la Santé et de la Recherche Médicale U1224, Paris, France.,Institut Pasteur, Microenvironnement and Immunity Unit, Paris, France
| | - Grégory Jouvion
- Institut Pasteur, Human Histopathology and Animal Models Unit, Paris, France
| | - Cindy Fevre
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Grégoire Gessain
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Pierre Thouvenot
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Gérard Eberl
- Institut National de la Santé et de la Recherche Médicale U1224, Paris, France.,Institut Pasteur, Microenvironnement and Immunity Unit, Paris, France
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1223, Paris, France
| | - Lucie Peduto
- Institut Pasteur, Stroma, Inflammation and Tissue Repair Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1224, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France .,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France.,Paris Descartes University, Department of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, APHP, Institut Imagine, Paris, France
| |
Collapse
|
50
|
Brand RM, Biswas N, Siegel A, Myerski A, Engstrom J, Jeffrey Metter E, Brand RE, Cranston RD, McGowan I. Immunological responsiveness of intestinal tissue explants and mucosal mononuclear cells to ex vivo stimulation. J Immunol Methods 2018; 463:39-46. [PMID: 30218652 DOI: 10.1016/j.jim.2018.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND There are limited data on the immunological responsiveness of healthy intestinal tissue when it is cultured and stimulated ex vivo. Such an ex vivo model has the potential to be a valuable tool in understanding disease pathogenesis and as a preclinical tool for the assessment of candidate therapeutic agents used to treat inflammatory bowel disease (IBD). AIM We undertook a comprehensive study to evaluate ex vivo immunological responses of intestinal tissue and isolated mucosal mononuclear cells (MMC) to a broad range of stimuli. METHODS Colorectal biopsies (explants) were obtained from healthy participants by flexible sigmoidoscopy and were placed either directly into culture or digested to isolate MMC prior to placement in culture. Explants or MMC were treated with polyinosinic:polycytidylic acid (Poly IC), phytohemagglutinin (PHA), lipopolysacccharides from E Coli (LPS), anti-CD3/CD28 antibodies, or IL-1β/TNF-α for 24 h. Supernatants were assayed for 40 inflammatory biomarkers using multiplexed enzyme-linked immunosorbent assay (ELISA). The isolated MMCs were further characterized using twelve color flow cytometry. RESULTS Explants have greater weight adjusted constitutive expression of inflammatory biomarkers than MMCs. Biomarker responses varied as a function of immunogen and use of intact tissue or isolated cells. PHA applied to intact explants was the most effective agent in inducing biomarker changes. Stimulation induced activated and memory cellular phenotypes in both explants and MMCs. CONCLUSIONS The breadth and magnitude of responses from intact and enzymatically digested intestinal tissue explants stimulated with exogenous immunogens are complex and vary by tissue form and treatment. Overall, PHA stimulation of intact explants produced the most robust responses in normal human colorectal tissue. This system could potentially serve as a preliminary model of the disease state, suitable for small scale screening of new therapeutic agents prior to using IBD patient derived tissue.
Collapse
Affiliation(s)
- Rhonda M Brand
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA.
| | - Nabanita Biswas
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aaron Siegel
- Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
| | - Ashley Myerski
- Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
| | - Jarret Engstrom
- Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
| | | | - Randall E Brand
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ross D Cranston
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ian McGowan
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
| |
Collapse
|