1
|
Wang ST, Yang Q, Liu MK, Li L, Wang W, Zhang SD, Zhang GF. Comparative transcriptomic analysis reveals a differential acid response mechanism between estuarine oyster (Crassostrea ariakensis) and Pacific oyster (Crassostrea gigas). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118210. [PMID: 40273612 DOI: 10.1016/j.ecoenv.2025.118210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/28/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Ocean and coastal acidification (OCA) poses a significant and rapidly emerging threat to mollusks. The physiological resilience of mollusks to OCA varies considerably; however, the underlying molecular mechanisms remain poorly understood. Seawater in estuaries, being more susceptible to acidification than that in open coastal zones, may enhance the tolerance of resident mollusks to low pH levels. Here, we conducted a comparative analysis between estuarine oysters (Crassostrea ariakensis) and Pacific oysters (Crassostrea gigas) using physiological phenotype and transcriptomic analyses to reveal differential acid-tolerance mechanisms in response to constant pH of 7.8. Our findings indicated that survival and respiration rates of C. ariakensis, which inhabits estuaries with fluctuating pH levels, were higher than those of C. gigas, which inhabits open coastal zones with relative stable pH conditions. Acid-responsive genes identified in C. gigas, including molecular chaperones and immune-related genes, exhibited higher constitutive expression in C. ariakensis under control conditions. Co-expression analyses revealed that C. ariakensis mitigated the effects of low pH by expressing genes involved in ion transporter activity and translation control. C. gigas activated genes associated with glycolipid metabolism while inhibiting cell division during acid stress. These findings suggested that C. ariakensis has evolved into a more energy-efficient regulatory network than C. gigas, incorporating both front-loading and responsive mechanisms to maintain acidbase homeostasis. This study is the first to investigate acid-tolerance differences between mollusks inhabiting estuarine and open coastal environments and provides critical insights into the resilience of mollusks in increasingly acidified oceans.
Collapse
Affiliation(s)
- Shen-Tong Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China
| | - Qi Yang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Ming-Kun Liu
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China; Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao, China
| | - Li Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China; Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao, China.
| | - Wei Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China
| | - Shou-Du Zhang
- Marine Science Research Institute of Shandong Province, Qingdao, China.
| | - Guo-Fan Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China; National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao 266071, China; Shandong Center of Technology Innovation for Oyster Seed Industry, Qingdao, China
| |
Collapse
|
2
|
Taylor H, Uhlig HH, Powrie F. Autoimmunity in inflammatory bowel disease: a holobiont perspective. Curr Opin Immunol 2025; 94:102557. [PMID: 40252635 DOI: 10.1016/j.coi.2025.102557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/21/2025]
Abstract
Adaptive immunity towards self-antigens (autoimmunity) and intestinal commensal microbiota is a key feature of inflammatory bowel disease (IBD). Considering mucosal adaptive immunity from a holobiont perspective, where the host and its microbiome form a single physiological unit, emphasises the challenge of avoiding damaging responses to self-antigen and symbiotic microbial communities in the gut while protecting against potential pathogens. Intestinal tolerance mechanisms prevent maladaptive T and B cell responses to microbial, environmental, and self-antigens, which drive inflammation. We discuss the spectrum of antimicrobial and autoantibody responses and highlight mechanisms by which common IBD-associated adaptive immune responses contribute to disease.
Collapse
Affiliation(s)
- Henry Taylor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Holm H Uhlig
- Centre for Human Genetics, University of Oxford, Oxford, UK; Translational Gastroenterology Liver Unit, University of Oxford, Oxford, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK; NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Chen Q, Gao Y, Li F, Yuan L. The role of gut-islet axis in pancreatic islet function and glucose homeostasis. Diabetes Obes Metab 2025; 27:1676-1692. [PMID: 39916498 PMCID: PMC11885102 DOI: 10.1111/dom.16225] [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: 09/03/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 03/08/2025]
Abstract
The gastrointestinal tract plays a vital role in the occurrence and treatment of metabolic diseases. Recent studies have convincingly demonstrated a bidirectional axis of communication between the gut and islets, enabling the gut to influence glucose metabolism and energy homeostasis in animals strongly. The 'gut-islet axis' is an essential endocrine signal axis that regulates islet function through the dialogue between intestinal microecology and endocrine metabolism. The discovery of glucagon-like peptide-1 (GLP-1), gastric inhibitory peptide (GIP) and other gut hormones has initially set up a bridge between gut and islet cells. However, the influence of other factors remains largely unknown, such as the homeostasis of the gut microbiota and the integrity of the gut barrier. Although gut microbiota primarily resides and affect intestinal function, they also affect extra-intestinal organs by absorbing and transferring metabolites derived from microorganisms. As a result of this transfer, islets may be continuously exposed to gut-derived metabolites and components. Changes in the composition of gut microbiota can damage the intestinal barrier function to varying degrees, resulting in increased intestinal permeability to bacteria and their derivatives. All these changes contribute to the severe disturbance of critical metabolic pathways in peripheral tissues and organs. In this review, we have outlined the different gut-islet axis signalling mechanisms associated with metabolism and summarized the latest progress in the complex signalling molecules of the gut and gut microbiota. In addition, we will discuss the impact of the gut renin-angiotensin system (RAS) on the various components of the gut-islet axis that regulate energy and glucose homeostasis. This work also indicates that therapeutic approaches aiming to restore gut microbial homeostasis, such as probiotics and faecal microbiota transplantation (FMT), have shown great potential in improving treatment outcomes, enhancing patient prognosis and slowing down disease progression. Future research should further uncover the molecular links between the gut-islet axis and the gut microbiota and explore individualized microbial treatment strategies, which will provide an innovative perspective and approach for the diagnosis and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Fangyu Li
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
4
|
Fukiage Y, Low S, Muramoto A, Ariga Y, Hoshino H, Nakada T, Akama TO, Kobayashi M. Expression of Glycoprotein 2 and Its Glycosylation in Human Cowper's Gland. J Histochem Cytochem 2025; 73:55-61. [PMID: 40026103 PMCID: PMC11683828 DOI: 10.1369/00221554241309413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/06/2024] [Indexed: 03/04/2025] Open
Abstract
Glycoprotein 2 (GP2), initially identified as a primary membrane protein of zymogen granules in pancreatic acinar cells, is a marker of intestinal microfold cells (M cells) and involved in bacterial transcytosis in M cells. Recent studies have reported GP2 expression in the pancreas and intestine among various other organs. We aimed to elucidate the expression of GP2 and its glycosylation modifications in human Cowper's glands. We generated an anti-human GP2 monoclonal antibody suitable for formalin-fixed, paraffin-embedded tissue sections. This antibody, designated GP2-71, successfully stained both the plasma membrane and cytoplasm of normal pancreatic acinar cells. Subsequently, we performed GP2-71 immunostaining on normal human Cowper's gland tissues. Our findings revealed that the luminal cell membrane and contents of the Cowper's glands reacted strongly with GP2-71. Moreover, the regions stained with GP2-71 were partially immunostained with the anti-sialyl Lewis x (sLex) monoclonal antibody HECA-452. Furthermore, western blot analysis of protein A-purified GP2-immunoglobulin G fusion proteins demonstrated that GP2 was decorated with HECA-452-positive glycans. Collectively, these findings indicate that GP2 is expressed in human Cowper's glands and is potentially decorated with sLex-related glycans.
Collapse
Affiliation(s)
- Yusuke Fukiage
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Shulin Low
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Akifumi Muramoto
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Yuzuru Ariga
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Hitomi Hoshino
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - Tsutomu Nakada
- Department of Instrumental Analysis, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto, Japan
| | - Tomoya O. Akama
- Department of Pharmacology, Kansai Medical University, Hirakata, Japan
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| |
Collapse
|
5
|
Xia CS, Krawczyk M, Di C, Krupa Ł, Kruk B, Krawczyk P, Milkiewicz P, Bao H, He X, Liu D, Fan C, Nasser A, Lopens S, Weiss FU, Frost F, Schierack P, Roggenbuck D, Liu Y. Loss of mucosal tolerance to glycoprotein 2 isoform 1 is a potential novel diagnostic biomarker for cholangiocarcinoma. Dig Liver Dis 2025; 57:14-22. [PMID: 38853092 DOI: 10.1016/j.dld.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/17/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Anti-glycoprotein 2 (anti-GP2) IgA and antineutrophil-cytoplasmic antibodies to proteinase 3 (PR3-ANCA) have been reported as predictive markers of cholangiocarcinoma (CCA) in patients with primary sclerosing cholangitis (PSC), but their prevalence in CCA patients without PSC remains unclear. METHODS This study involved Asian discovery (n = 118) and European validation (n = 38) cohorts of CCA patients without PSC, alongside 49 Asian and 82 European pancreatic ductal adenocarcinoma (PDAC) patients, 21 with benign pancreatic neoplasms (BPN) and 45 with hepatocellular carcinoma (HCC), and 157 healthy controls (HC) from Asia and Europe. We analyzed the prevalence of PR3-ANCA, IgA and IgG against GP21 and GP24, and the CA19-9 levels. RESULTS Anti-GP21 IgA was the most prevalent in both CCA cohorts (discovery: 55.1 %; validation: 42.1 %) and significantly higher than in other groups except PDAC (all p < 0.05). It demonstrated the best diagnostic performance in distinguishing CCA from disease controls and HC, outperforming tumor markers. No significant correlation was found between anti-GP21 IgA levels and CA19-9 levels. CONCLUSION Our findings show that anti-GP21 IgA revealing the loss of mucosal tolerance is a potential novel diagnostic biomarker for CCA.
Collapse
Affiliation(s)
- Chang-Sheng Xia
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany; Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Chun Di
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Łukasz Krupa
- Department of Gastroenterology and Hepatology with Internal Disease Unit, Teaching Hospital No 1 in Rzeszów, Medical Department, University of Rzeszów, Poland
| | - Beata Kruk
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Krawczyk
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Poland
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; Translational Medicine Group, Pomeranian Medical University, Szczecin, Poland
| | - Huizhang Bao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiao He
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Damin Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chunhong Fan
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | | | - Steffi Lopens
- Medipan GmbH, R/D Department, Dahlewitz, Germany; Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Fabian Frost
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Peter Schierack
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Dirk Roggenbuck
- Medipan GmbH, R/D Department, Dahlewitz, Germany; Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Yudong Liu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing 100730, PR China.
| |
Collapse
|
6
|
Schmidt HF, Darwin CB, Sundaram MV. The Pax transcription factor EGL-38 links EGFR signaling to assembly of a cell type-specific apical extracellular matrix in the Caenorhabditis elegans vulva. Dev Biol 2025; 517:265-277. [PMID: 39489317 PMCID: PMC11631643 DOI: 10.1016/j.ydbio.2024.10.008] [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: 09/05/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The surface of epithelial tissues is covered by an apical extracellular matrix (aECM). The aECMs of different tissues have distinct compositions to serve distinct functions, yet how a particular cell type assembles the proper aECM is not well understood. We used the cell type-specific matrix of the C. elegans vulva to investigate the connection between cell identity and matrix assembly. The vulva is an epithelial tube composed of seven cell types descending from EGFR/Ras-dependent (1°) and Notch-dependent (2°) lineages. Vulva aECM contains multiple Zona Pellucida domain (ZP) proteins, which are a common component of aECMs across life. ZP proteins LET-653 and CUTL-18 assemble on 1° cell surfaces, while NOAH-1 assembles on a subset of 2° surfaces. All three ZP genes are broadly transcribed, indicating that cell type-specific ZP assembly must be determined by features of the destination cell surface. The paired box (Pax) transcription factor EGL-38 promotes assembly of 1° matrix and prevents inappropriate assembly of 2° matrix, suggesting that EGL-38 promotes expression of one or more ZP matrix organizers. Our results connect the known signaling pathways and various downstream effectors to EGL-38/Pax expression and the ZP matrix component of vulva cell fate execution. We propose that dedicated transcriptional networks may contribute to cell-appropriate assembly of aECM in many epithelial organs.
Collapse
Affiliation(s)
- Helen F Schmidt
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Chelsea B Darwin
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Zhou Y, Komnick MR, Sepulveda F, Liu G, Nieves-Ortiz E, Meador K, Ndatabaye O, Fatkhullina A, Wu-Woods NJ, Naydenkov PM, Kent J, Christiansen N, Madariaga ML, Witkowski P, Ismagilov RF, Esterházy D. Inducible, but not constitutive, pancreatic REG/Reg isoforms are regulated by intestinal microbiota and pancreatic diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619139. [PMID: 39484594 PMCID: PMC11526982 DOI: 10.1101/2024.10.18.619139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The REG / Reg gene locus encodes for a conserved family of potent antimicrobial but also pancreatitis-associated proteins. Here we investigated whether REG/Reg family members differ in their baseline expression levels and abilities to be regulated in the pancreas and gut upon perturbations. We found, in human and mouse, pancreas and gut differed in REG / Reg isoform levels and preferences, with duodenum most resembling the pancreas. Pancreatic acinar cells and intestinal enterocytes were the dominant REG producers. Intestinal symbiotic microbes regulated the expression of the same, select Reg members in gut and pancreas. These Reg members had the most STAT3-binding sites close to the transcription start sites and were partially IL-22 dependent. We thus categorized them as "inducible" and others as "constitutive". Indeed, also in models of pancreatic-ductal adenocarcinoma and pancreatitis, only inducible Reg members were upregulated in pancreas. While intestinal Reg expression remained unchanged upon pancreatic perturbation, pancreatitis altered the microbial composition of the duodenum and feces shortly after disease onset. Our study reveals differential usage and regulation of REG / Reg isoforms as a mechanism for tissue-specific innate immunity, highlights the intimate connection of pancreas and duodenum, and implies a gut-to-pancreas communication axis resulting in a coordinated Reg response.
Collapse
|
8
|
Frost F, Weiss S, Hertel J, Rühlemann M, Bang C, Franke A, Nauck M, Dörr M, Völzke H, Roggenbuck D, Schierack P, Völker U, Homuth G, Aghdassi AA, Sendler M, Lerch MM, Weiss FU. Fecal glycoprotein 2 is a marker of gut microbiota dysbiosis and systemic inflammation. Gut Pathog 2024; 16:60. [PMID: 39427219 PMCID: PMC11490104 DOI: 10.1186/s13099-024-00657-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Antimicrobial autoantigenic glycoprotein 2 (GP2) is an important component of the innate immune system which originates from the exocrine pancreas as well as from the small intestines. The relationship of GP2 with the intestinal microbiome as well as the systemic implications of increased fecal GP2 levels are, however, still unclear. Therefore, fecal samples from 2,812 individuals of the Study of Health in Pomerania (SHIP) were collected to determine GP2 levels (enzyme-linked immunosorbent assay) and gut microbiota profiles (16 S rRNA gene sequencing). These data were correlated and associated with highly standardised and comprehensive phenotypic data of the study participants. RESULTS Fecal GP2 levels were increased in individuals with higher body mass index and smokers, whereas lower levels were found in case of preserved exocrine pancreatic function, female sex or a healthier diet. Moreover, higher GP2 levels were associated with increased serum levels of high-sensitivity C-reactive protein, loss of gut microbial diversity and an increase of potentially detrimental bacteria (Streptococcus, Haemophilus, Clostridium XIVa, or Collinsella). At the same time, predicted microbial pathways for the biosynthesis of beneficial short-chain fatty acids or lactic acid were depleted in individuals with high fecal GP2. Of note, GP2 exhibited a stronger association to overall microbiome variation than calprotectin. CONCLUSION Fecal GP2 is a biomarker of gut microbiota dysbiosis and associated with increased systemic inflammation. The intestines may be more important as origin for GP2 than pancreatic acinar cells. Future studies need to investigate the potential clinical value in disease specific patient cohorts.
Collapse
Affiliation(s)
- Fabian Frost
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch- Straße, 17475, Greifswald, Germany
| | - Stefan Weiss
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch- Straße, 17475, Greifswald, Germany
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Johannes Hertel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Malte Rühlemann
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Dirk Roggenbuck
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Peter Schierack
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Ali A Aghdassi
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch- Straße, 17475, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch- Straße, 17475, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch- Straße, 17475, Greifswald, Germany
| | - Frank U Weiss
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch- Straße, 17475, Greifswald, Germany.
| |
Collapse
|
9
|
Shay TF, Jang S, Brittain TJ, Chen X, Walker B, Tebbutt C, Fan Y, Wolfe DA, Arokiaraj CM, Sullivan EE, Ding X, Wang TY, Lei Y, Chuapoco MR, Chou TF, Gradinaru V. Human cell surface-AAV interactomes identify LRP6 as blood-brain barrier transcytosis receptor and immune cytokine IL3 as AAV9 binder. Nat Commun 2024; 15:7853. [PMID: 39245720 PMCID: PMC11381518 DOI: 10.1038/s41467-024-52149-0] [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: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
Adeno-associated viruses (AAVs) are foundational gene delivery tools for basic science and clinical therapeutics. However, lack of mechanistic insight, especially for engineered vectors created by directed evolution, can hamper their application. Here, we adapt an unbiased human cell microarray platform to determine the extracellular and cell surface interactomes of natural and engineered AAVs. We identify a naturally-evolved and serotype-specific interaction between the AAV9 capsid and human interleukin 3 (IL3), with possible roles in host immune modulation, as well as lab-evolved low-density lipoprotein receptor-related protein 6 (LRP6) interactions specific to engineered capsids with enhanced blood-brain barrier crossing in non-human primates after intravenous administration. The unbiased cell microarray screening approach also allows us to identify off-target tissue binding interactions of engineered brain-enriched AAV capsids that may inform vectors' peripheral organ tropism and side effects. Our cryo-electron tomography and AlphaFold modeling of capsid-interactor complexes reveal LRP6 and IL3 binding sites. These results allow confident application of engineered AAVs in diverse organisms and unlock future target-informed engineering of improved viral and non-viral vectors for non-invasive therapeutic delivery to the brain.
Collapse
Affiliation(s)
- Timothy F Shay
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Seongmin Jang
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tyler J Brittain
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xinhong Chen
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Beth Walker
- Charles River Laboratories, High Peak Business Park, Buxton Road, Chinley, SK23 6FJ, UK
| | - Claire Tebbutt
- Charles River Laboratories, High Peak Business Park, Buxton Road, Chinley, SK23 6FJ, UK
| | - Yujie Fan
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Damien A Wolfe
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Cynthia M Arokiaraj
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Erin E Sullivan
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xiaozhe Ding
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Ting-Yu Wang
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Yaping Lei
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Miguel R Chuapoco
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tsui-Fen Chou
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Viviana Gradinaru
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
10
|
Schmidt HF, Darwin CB, Sundaram MV. The Pax transcription factor EGL-38 links EGFR signaling to assembly of a cell-type specific apical extracellular matrix in the Caenorhabditis elegans vulva. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611291. [PMID: 39282387 PMCID: PMC11398461 DOI: 10.1101/2024.09.04.611291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The surface of epithelial tissues is covered by an apical extracellular matrix (aECM). The aECMs of different tissues have distinct compositions to serve distinct functions, yet how a particular cell type assembles the proper aECM is not well understood. We used the cell-type specific matrix of the C. elegans vulva to investigate the connection between cell identity and matrix assembly. The vulva is an epithelial tube composed of seven cell types descending from EGFR/Ras-dependent (1°) and Notch-dependent (2°) lineages. Vulva aECM contains multiple Zona Pellucida domain (ZP) proteins, which are a common component of aECMs across life. ZP proteins LET-653 and CUTL-18 assemble on 1° cell surfaces, while NOAH-1 assembles on a subset of 2° surfaces. All three ZP genes are broadly transcribed, indicating that cell-type specific ZP assembly must be determined by features of the destination cell surface. The paired box (Pax) transcription factor EGL-38 promotes assembly of 1° matrix and prevents inappropriate assembly of 2° matrix, suggesting that EGL-38 promotes expression of one or more ZP matrix organizers. Our results connect the known signaling pathways and various downstream effectors to EGL-38/Pax expression and the ZP matrix component of vulva cell fate execution. We propose that dedicated transcriptional networks may contribute to cell-appropriate assembly of aECM in many epithelial organs.
Collapse
Affiliation(s)
- Helen F Schmidt
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chelsea B Darwin
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Lopens S, Schierack P, Krause J, Piaszczyński M, Król R, Staroń R, Krupa Ł, Gutkowski K, Kruk B, Grąt M, Krawczyk M, Patkowski W, Glaser F, Rödiger S, Grossmann K, Pająk J, Milkiewicz P, Lammert F, Zieniewicz K, Schramm C, Roggenbuck D, Krawczyk M. Antimicrobial glycoprotein 2 (GP2) in gallstones, bile fluid and peribiliary glands of patients with primary sclerosing cholangitis. Clin Chim Acta 2024; 562:119841. [PMID: 38964568 DOI: 10.1016/j.cca.2024.119841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 05/28/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Glycoprotein-2 (GP2) IgA is a predictor of disease severity in primary sclerosing cholangitis (PSC). We examined GP2's occurrence in the biliary tract, the site of inflammation. METHODS GP2 was analyzed using ELISA, immunoblotting, mass spectrometry, and immunohistochemistry. The samples included: 20 bile and 30 serum samples from PSC patients, 23 bile and 11 serum samples from patients with gallstone disease (GD), 15 bile samples from healthy individuals undergoing liver-donation surgery (HILD), 20 extracts of gallstones (GE) obtained during cholecystectomy, and 101 blood-donor sera. RESULTS Biliary GP2 concentrations were significantly higher in patients with PSC and GD than in HILD (p < 0.0001). Serum GP2 levels were similar in PSC and GD patients, and controls, but lower than in bile (p < 0.0001). GP2 was detected in all 20 GEs. Mass spectrometry identified GP2 in the bile of 2 randomly selected GD and 2 PSC patients, and in none of 2 HILD samples. GP2 was found in peribiliary glands in 8 out of 12 PSC patients, showing morphological changes in acinar cells, but not in GD-gallbladders. CONCLUSIONS GP2 is present in bile of PSC and GD patients. It is synthesized in the peribiliary glands of PSC patients, supporting a pathogenic role for biliary GP2 in PSC.
Collapse
Affiliation(s)
- Steffi Lopens
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Medipan GmbH, Dahlewitz, Germany
| | - Peter Schierack
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Jenny Krause
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michał Piaszczyński
- Department of General, Vascular and Transplant Surgery, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Robert Król
- Department of General, Vascular and Transplant Surgery, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Robert Staroń
- Department of Gastroenterology and Hepatology with Internal Disease Unit, Teaching Hospital No 1 in Rzeszów, Rzeszów, Poland; Medical Department, University of Rzeszów, Rzeszów, Poland
| | - Łukasz Krupa
- Department of Gastroenterology and Hepatology with Internal Disease Unit, Teaching Hospital No 1 in Rzeszów, Rzeszów, Poland; Medical Department, University of Rzeszów, Rzeszów, Poland
| | - Krzysztof Gutkowski
- Department of Gastroenterology and Hepatology with Internal Disease Unit, Teaching Hospital No 1 in Rzeszów, Rzeszów, Poland; Medical Department, University of Rzeszów, Rzeszów, Poland
| | - Beata Kruk
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Michał Grąt
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Marek Krawczyk
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Waldemar Patkowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Fabian Glaser
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rödiger
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | | | - Jacek Pająk
- Department of Pathomorphology and Molecular Diagnostics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland; Translational Medicine Group, Pomeranian Medical University, Szczecin, Poland
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany; Health Sciences, Hannover Medical School (MHH), Hannover, Germany
| | - Krzysztof Zieniewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Christoph Schramm
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Roggenbuck
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany; Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany.
| | - Marcin Krawczyk
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
12
|
Inoue Y, Uemura Y, Kosugi S, Kanno M, Sano F. Unique Pharmacokinetics for Oral Tacrolimus Administration After Allogeneic Hematopoietic Stem-Cell Transplantation for Acute Myeloid Leukemia With Shwachman-Diamond Syndrome. Am J Ther 2024; 31:e318-e319. [PMID: 37831700 DOI: 10.1097/mjt.0000000000001639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Affiliation(s)
- Yasuyuki Inoue
- Division of Hematology, Department of Internal Medicine, St. Marianna University Yokohama-city Seibu Hospital, Yokohama Japan; and
| | - Yu Uemura
- Division of Hematology, Department of Internal Medicine, St. Marianna University, Kawasaki Japan
| | - Shigeki Kosugi
- Division of Hematology, Department of Internal Medicine, St. Marianna University Yokohama-city Seibu Hospital, Yokohama Japan; and
| | - Masatoshi Kanno
- Division of Hematology, Department of Internal Medicine, St. Marianna University Yokohama-city Seibu Hospital, Yokohama Japan; and
| | - Fumiaki Sano
- Division of Hematology, Department of Internal Medicine, St. Marianna University, Kawasaki Japan
| |
Collapse
|
13
|
Lopez-Gordo E, Chamberlain K, Riyad JM, Kohlbrenner E, Weber T. Natural Adeno-Associated Virus Serotypes and Engineered Adeno-Associated Virus Capsid Variants: Tropism Differences and Mechanistic Insights. Viruses 2024; 16:442. [PMID: 38543807 PMCID: PMC10975205 DOI: 10.3390/v16030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
Today, adeno-associated virus (AAV)-based vectors are arguably the most promising in vivo gene delivery vehicles for durable therapeutic gene expression. Advances in molecular engineering, high-throughput screening platforms, and computational techniques have resulted in a toolbox of capsid variants with enhanced performance over parental serotypes. Despite their considerable promise and emerging clinical success, there are still obstacles hindering their broader use, including limited transduction capabilities, tissue/cell type-specific tropism and penetration into tissues through anatomical barriers, off-target tissue biodistribution, intracellular degradation, immune recognition, and a lack of translatability from preclinical models to clinical settings. Here, we first describe the transduction mechanisms of natural AAV serotypes and explore the current understanding of the systemic and cellular hurdles to efficient transduction. We then outline progress in developing designer AAV capsid variants, highlighting the seminal discoveries of variants which can transduce the central nervous system upon systemic administration, and, to a lesser extent, discuss the targeting of the peripheral nervous system, eye, ear, lung, liver, heart, and skeletal muscle, emphasizing their tissue and cell specificity and translational promise. In particular, we dive deeper into the molecular mechanisms behind their enhanced properties, with a focus on their engagement with host cell receptors previously inaccessible to natural AAV serotypes. Finally, we summarize the main findings of our review and discuss future directions.
Collapse
|
14
|
Sendid B, Cornu M, Cordier C, Bouckaert J, Colombel JF, Poulain D. From ASCA breakthrough in Crohn's disease and Candida albicans research to thirty years of investigations about their meaning in human health. Autoimmun Rev 2024; 23:103486. [PMID: 38040100 DOI: 10.1016/j.autrev.2023.103486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Anti-Saccharomyces cerevisiae antibodies (ASCA) are human antibodies that can be detected using an enzyme-linked immunosorbent assay involving a mannose polymer (mannan) extracted from the cell wall of the yeast S. cerevisiae. The ASCA test was developed in 1993 with the aim of differentiating the serological response in two forms of inflammatory bowel disease (IBD), Crohn's disease and ulcerative colitis. The test, which is based on the detection of anti-oligomannosidic antibodies, has been extensively performed worldwide and there have been hundreds of publications on ASCA. The earlier studies concerned the initial diagnostic indications of ASCA and investigations then extended to many human diseases, generally in association with studies on intestinal microorganisms and the interaction of the micro-mycobiome with the immune system. The more information accumulates, the more the mystery of the meaning of ASCA deepens. Many fundamental questions remain unanswered. These questions concern the heterogeneity of ASCA, the mechanisms of their generation and persistence, the existence of self-antigens, and the relationship between ASCA and inflammation and autoimmunity. This review aims to discuss the gray areas concerning the origin of ASCA from an analysis of the literature. Structured around glycobiology and the mannosylated antigens of S. cerevisiae and Candida albicans, this review will address these questions and will try to clarify some lines of thought. The importance of the questions relating to the pathophysiological significance of ASCA goes far beyond IBD, even though these diseases remain the preferred models for their understanding.
Collapse
Affiliation(s)
- Boualem Sendid
- INSERM U1285, CNRS UMR 8576, Glycobiology in Fungal Pathogenesis and Clinical Applications, Université de Lille, F-59000 Lille, France; Pôle de Biologie-Pathologie-Génétique, Institut de Microbiologie, Service de Parasitologie-Mycologie, CHU Lille, F-59000 Lille, France.
| | - Marjorie Cornu
- INSERM U1285, CNRS UMR 8576, Glycobiology in Fungal Pathogenesis and Clinical Applications, Université de Lille, F-59000 Lille, France; Pôle de Biologie-Pathologie-Génétique, Institut de Microbiologie, Service de Parasitologie-Mycologie, CHU Lille, F-59000 Lille, France
| | - Camille Cordier
- INSERM U1285, CNRS UMR 8576, Glycobiology in Fungal Pathogenesis and Clinical Applications, Université de Lille, F-59000 Lille, France; Pôle de Biologie-Pathologie-Génétique, Institut de Microbiologie, Service de Parasitologie-Mycologie, CHU Lille, F-59000 Lille, France
| | - Julie Bouckaert
- CNRS UMR 8576, Computational Molecular Systems Biology, Université de Lille, F-59000 Lille, France
| | - Jean Frederic Colombel
- Department of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel Poulain
- INSERM U1285, CNRS UMR 8576, Glycobiology in Fungal Pathogenesis and Clinical Applications, Université de Lille, F-59000 Lille, France.
| |
Collapse
|
15
|
Zhang Z, Tanaka I, Nakahashi-Ouchida R, Ernst PB, Kiyono H, Kurashima Y. Glycoprotein 2 as a gut gate keeper for mucosal equilibrium between inflammation and immunity. Semin Immunopathol 2024; 45:493-507. [PMID: 38170255 PMCID: PMC11136868 DOI: 10.1007/s00281-023-00999-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Glycoprotein 2 (GP2) is a widely distributed protein in the digestive tract, contributing to mucosal barrier maintenance, immune homeostasis, and antigen-specific immune response, while also being linked to inflammatory bowel disease (IBD) pathogenesis. This review sheds light on the extensive distribution of GP2 within the gastrointestinal tract and its intricate interplay with the immune system. Furthermore, the significance of GP2 autoantibodies in diagnosing and categorizing IBD is underscored, alongside the promising therapeutic avenues for modulating GP2 to regulate immunity and maintain mucosal balance.
Collapse
Affiliation(s)
- Zhongwei Zhang
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
- Chiba University Futuristic Mucosal Vaccine Research and Development Synergy Institute (cSIMVa), Chiba, Japan
| | - Izumi Tanaka
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
- Chiba University Futuristic Mucosal Vaccine Research and Development Synergy Institute (cSIMVa), Chiba, Japan
| | - Rika Nakahashi-Ouchida
- Chiba University Futuristic Mucosal Vaccine Research and Development Synergy Institute (cSIMVa), Chiba, Japan
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Peter B Ernst
- Department of Medicine, School of Medicine, Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), San Diego, CA, USA
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, CA, USA
- Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan
| | - Hiroshi Kiyono
- Chiba University Futuristic Mucosal Vaccine Research and Development Synergy Institute (cSIMVa), Chiba, Japan
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Department of Medicine, School of Medicine, Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), San Diego, CA, USA
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan
- HanaVax Inc., Tokyo, Japan
- Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
- Chiba University Futuristic Mucosal Vaccine Research and Development Synergy Institute (cSIMVa), Chiba, Japan.
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan.
- Department of Medicine, School of Medicine, Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), San Diego, CA, USA.
- Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan.
| |
Collapse
|
16
|
Morisseau L, Tokito F, Poulain S, Plaisance V, Pawlowski V, Kim SH, Legallais C, Jellali R, Sakai Y, Abderrahmani A, Leclerc E. Generation of β-like cell subtypes from differentiated human induced pluripotent stem cells in 3D spheroids. Mol Omics 2023; 19:810-822. [PMID: 37698079 DOI: 10.1039/d3mo00050h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Since the identification of four different pancreatic β-cell subtypes and bi-hormomal cells playing a role in the diabetes pathogenesis, the search for in vitro models that mimics such cells heterogeneity became a key priority in experimental and clinical diabetology. We investigated the potential of human induced pluripotent stem cells to lead to the development of the different β-cells subtypes in honeycomb microwell-based 3D spheroids. The glucose-stimulated insulin secretion confirmed the spheroids functionality. Then, we performed a single cell RNA sequencing of the spheroids. Using a knowledge-based analysis with a stringency on the pancreatic markers, we extracted the β-cells INS+/UCN3+ subtype (11%; β1-like cells), the INS+/ST8SIA1+/CD9- subtype (3%, β3-like cells) and INS+/CD9+/ST8SIA1-subtype (1%; β2-like cells) consistently with literature findings. We did not detect the INS+/ST8SIA1+/CD9+ cells (β4-like cells). Then, we also identified four bi-hormonal cells subpopulations including δ-like cells (INS+/SST+, 6%), γ-like cells (INS+/PPY+, 3%), α-like-cells (INS+/GCG+, 6%) and ε-like-cells (INS+/GHRL+, 2%). Using data-driven clustering, we extracted four progenitors' subpopulations (with the lower level of INS gene) that included one population highly expressing inhibin genes (INHBA+/INHBB+), one population highly expressing KCNJ3+/TPH1+, one population expressing hepatocyte-like lineage markers (HNF1A+/AFP+), and one population expressing stem-like cell pancreatic progenitor markers (SOX2+/NEUROG3+). Furthermore, among the cycling population we found a large number of REST+ cells and CD9+ cells (CD9+/SPARC+/REST+). Our data confirm that our differentiation leads to large β-cell heterogeneity, which can be used for investigating β-cells plasticity under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Lisa Morisseau
- Biomechanics and Bioengineering UMR 7338, Université de technologie de Compiègne, CNRS, Centre de Recherche Royallieu CS 60319, Compiègne, 60203 Cedex, France
| | - Fumiya Tokito
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Stéphane Poulain
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| | - Valerie Plaisance
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Valerie Pawlowski
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Soo Hyeon Kim
- Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| | - Cécile Legallais
- Biomechanics and Bioengineering UMR 7338, Université de technologie de Compiègne, CNRS, Centre de Recherche Royallieu CS 60319, Compiègne, 60203 Cedex, France
| | - Rachid Jellali
- Biomechanics and Bioengineering UMR 7338, Université de technologie de Compiègne, CNRS, Centre de Recherche Royallieu CS 60319, Compiègne, 60203 Cedex, France
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
- Laboratory for Integrated Micro Mechatronic Systems, CNRS/IIS IRL 2820, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| | - Amar Abderrahmani
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520, IEMN, F-59000 Lille, France
| | - Eric Leclerc
- Laboratory for Integrated Micro Mechatronic Systems, CNRS/IIS IRL 2820, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba; Meguro-ku, Tokyo, 153-8505, Japan
| |
Collapse
|
17
|
Kitamoto S, Kamada N. The oral-gut axis: a missing piece in the IBD puzzle. Inflamm Regen 2023; 43:54. [PMID: 37932859 PMCID: PMC10626704 DOI: 10.1186/s41232-023-00304-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial intractable intestinal disease. Focusing on only one facet of the pathogenesis of IBD is insufficient to fully capture the complexity of the disease, and results in limited advance in clinical management. Therefore, it is critical to dissect the interactions amongst the multifarious contributors to the pathogenesis to comprehensively understand its pathology and subsequently improve clinical outcomes. In this context, the systemic interactions between organs, particularly the oral-gut axis mediated by host immune cells and resident microorganisms, have garnered significant attention in IBD research. More specifically, periodontal disease such as periodontitis has been implicated in augmenting intestinal inflammation beyond the confines of the oral cavity. There is mounting evidence suggesting that potentially harmful oral resident bacteria, termed pathobionts, and pro-inflammatory immune cells from the oral mucosa can migrate to the gastrointestinal tract, thereby potentiating intestinal inflammation. This article aims to provide a holistic overview of the causal relationship between periodontal disease and intestinal inflammation. Furthermore, we will discuss potential determinants that facilitate the translocation of oral pathobionts into the gut, a key event underpinning the oral-gut axis. Unraveling the complex dynamics of microbiota and immunity in the oral-gut continuum will lead to a better understanding of the pathophysiology inherent in both oral and intestinal diseases and the development of prospective therapeutic strategies.
Collapse
Affiliation(s)
- Sho Kitamoto
- The World Premier International Research Center (WPI) Immunology Frontier Research Center (IFReC), 1012 IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Nobuhiko Kamada
- The World Premier International Research Center (WPI) Immunology Frontier Research Center (IFReC), 1012 IFReC Research Building, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI, 48109, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
18
|
Abaidullah M, La S, Liu M, Liu B, Cui Y, Wang Z, Sun H, Ma S, Shi Y. Polysaccharide from Smilax glabra Roxb Mitigates Intestinal Mucosal Damage by Therapeutically Restoring the Interactions between Gut Microbiota and Innate Immune Functions. Nutrients 2023; 15:4102. [PMID: 37836386 PMCID: PMC10574425 DOI: 10.3390/nu15194102] [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: 08/27/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023] Open
Abstract
Smilax glabra Roxb (S. glabra) is a conventional Chinese medicine that is mainly used for the reliability of inflammation. However, bioactive polysaccharides from S. glabra (SGPs) have not been thoroughly investigated. Here, we demonstrate for the first time that SGPs preserve the integrity of the gut epithelial layer and protect against intestinal mucosal injury induced by dextran sulfate sodium. Mechanistically, SGPs mitigated colonic mucosal injury by restoring the association between the gut flora and innate immune functions. In particular, SGPs increased the number of goblet cells, reduced the proportion of apoptotic cells, improved the differentiation of gut tight junction proteins, and enhanced mucin production in the gut epithelial layer. Moreover, SGPs endorsed the propagation of probiotic bacteria, including Lachnospiraceae bacterium, which strongly correlated with decreased pro-inflammatory cytokines via the blocking of the TLR-4 NF-κB and MyD88 pathways. Overall, our study establishes a novel use of SGPs for the treatment of inflammatory bowel disease (IBD)-associated mucosal injury and provides a basis for understanding the therapeutic effects of natural polysaccharides from the perspective of symbiotic associations between host innate immune mechanisms and the gut microbiome.
Collapse
Affiliation(s)
- Muhammad Abaidullah
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Shaokai La
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
| | - Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yalei Cui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Hao Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Sen Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| |
Collapse
|
19
|
Huang Y, Jonsson NN, McLaughlin M, Burchmore R, Johnson PCD, Jones RO, McGill S, Brady N, Weidt S, Eckersall PD. Quantitative TMT-based proteomics revealing host, dietary and microbial proteins in bovine faeces including barley serpin Z4, a prominent component in the head of beer. J Proteomics 2023; 285:104941. [PMID: 37285906 DOI: 10.1016/j.jprot.2023.104941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
There has been little information about the proteome of bovine faeces or about the contribution to the faecal proteome of proteins from the host, the feed or the intestinal microbiome. Here, the bovine faecal proteome and the origin of its component proteins was assessed, while also determining the effect of treating barley, the major carbohydrate in the feed, with either ammonia (ATB) or sodium propionate (PTB) preservative. Healthy continental crossbreed steers were allocated to two groups and fed on either of the barley-based diets. Five faecal samples from each group were collected on Day 81 of the trial and analysed by quantitative proteomics using nLC-ESI-MS/MS after tandem mass tag labelling. In total, 281 bovine proteins, 199 barley proteins, 176 bacterial proteins and 190 archaeal proteins were identified in the faeces. Mucosal pentraxin, albumin and digestive enzymes were among bovine proteins identified. Serpin Z4 a protease inhibitor was the most abundant barley protein identified which is also found in barley-based beer, while numerous microbial proteins were identified, many originating bacteria from Clostridium, while Methanobrevibacter was the dominant archaeal genus. Thirty-nine proteins were differentially abundant between groups, the majority being more abundant in the PTB group compared to the ATB group. SIGNIFICANCE: Proteomic examination of faeces is becoming a valuable means to assess the health of the gastro-intestinal tract in several species, but knowledge on the proteins present in bovine faeces is limited. This investigation aimed to characterise the proteome of bovine faecal extracts in order to evaluate the potential for investigations of the proteome as a means to assess the health, disease and welfare of cattle in the future. The investigation was able to identify proteins in bovine faeces that had been (i) produced by the individual cattle, (ii) present in the barley-based feed eaten by the cattle or (iii) produced by bacteria and other microbes in the rumen or intestines. Bovine proteins identified included mucosal pentraxin, serum albumin and a variety of digestive enzymes. Barley proteins found in the faeces included serpin Z4, a protease inhibitor that is also found in beer having survived the brewing process. Bacterial and archaeal proteins in the faecal extracts were related to several pathways related to the metabolism of carbohydrates. The recognition of the range of proteins that can be identified in bovine faeces raises the possibility that non-invasive sample collection of this material could provide a novel diagnostic approach to cattle health and welfare.
Collapse
Affiliation(s)
- Y Huang
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK
| | - N N Jonsson
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK
| | - M McLaughlin
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK
| | - R Burchmore
- Institute of Infection, Immunity & Inflammation and Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - P C D Johnson
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK
| | - R O Jones
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK
| | - S McGill
- Institute of Infection, Immunity & Inflammation and Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - N Brady
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK
| | - S Weidt
- Institute of Infection, Immunity & Inflammation and Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - P D Eckersall
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, G61 1QH, UK; Interdisciplinary Laboratory of Clinical Analysis of the University of Murcia (Interlab-UMU), Department of Animal Medicine and Surgery, Veterinary School, University of Murcia, Murcia 30100, Spain.
| |
Collapse
|
20
|
Daniluk U, Krawiec P, Pac-Kożuchowska E, Dembiński Ł, Bukowski JS, Banaszkiewicz A, Woźniuk-Kaźmierczak A, Czkwianianc E, Brylak J, Walkowiak J, Borys-Iwanicka A, Kofla-Dłubacz A, Pytrus T, Zdanowicz K, Lebensztejn DM. Pancreatic Involvement in the Course of Inflammatory Bowel Disease in Children-A Multi-Center Study. J Clin Med 2023; 12:4174. [PMID: 37445210 DOI: 10.3390/jcm12134174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
The coexistence of inflammatory bowel disease (IBD) with pancreatic pathology is rare in children. A retrospective analysis of data from 1538 children diagnosed with IBD in 2014-2021 was conducted to determine the frequency and causes of pancreatitis and asymptomatic hyperlipasemia (HL) or hyperamylasemia (HA) in this group of patients. Among the 176 children (11.4%) with pancreatic involvement (PI), acute pancreatitis (AP) was diagnosed in 77 children (43.8%), and HA or HL was observed in 88 children (50.0%). Only a few patients were diagnosed with autoimmune or chronic pancreatitis (6.2%). PI was observed at the time of the IBD diagnosis in 26.1% of the cases. A total of 54.5% of the patients had moderate to severe IBD, and 96% had colonic involvement at the time of diagnosis of PI. Idiopathic PI was the most common (57%), followed by drug-induced PI (37%) and azathioprine (AZA). In patients with AZA-induced AP, the successful introduction of 6-mercaptopurine (6-MP) to therapy was noted in 62.5% of the children. Our results suggest that routine monitoring of pancreatic enzymes in patients with IBD should be performed, especially after the initiation of the AZA treatment. The presence of transient HA/HL in IBD does not necessarily indicate pancreatic pathology.
Collapse
Affiliation(s)
- Urszula Daniluk
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition, Allergology and Pulmonology, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Paulina Krawiec
- Department of Pediatrics and Gastroenterology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Elżbieta Pac-Kożuchowska
- Department of Pediatrics and Gastroenterology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Łukasz Dembiński
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Jan Stanisław Bukowski
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Aleksandra Banaszkiewicz
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Woźniuk-Kaźmierczak
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Elżbieta Czkwianianc
- Department of Gastroenterology, Allergology and Pediatrics, Polish Mother's Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Jan Brylak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 60-572 Poznan, Poland
| | - Agnieszka Borys-Iwanicka
- 2nd Clinical Department of Pediatrics, Gastroenterology and Nutrition, Medical University of Wroclaw, 50-369 Wroclaw, Poland
| | - Anna Kofla-Dłubacz
- 2nd Clinical Department of Pediatrics, Gastroenterology and Nutrition, Medical University of Wroclaw, 50-369 Wroclaw, Poland
| | - Tomasz Pytrus
- 2nd Clinical Department of Pediatrics, Gastroenterology and Nutrition, Medical University of Wroclaw, 50-369 Wroclaw, Poland
| | - Katarzyna Zdanowicz
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition, Allergology and Pulmonology, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Dariusz Marek Lebensztejn
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition, Allergology and Pulmonology, Medical University of Bialystok, 15-274 Bialystok, Poland
| |
Collapse
|
21
|
Morino K, Kunimura K, Sugiura Y, Izumi Y, Matsubara K, Akiyoshi S, Maeda R, Hirotani K, Sakata D, Mizuno S, Takahashi S, Bamba T, Uruno T, Fukui Y. Cholesterol sulfate limits neutrophil recruitment and gut inflammation during mucosal injury. Front Immunol 2023; 14:1131146. [PMID: 37006281 PMCID: PMC10063914 DOI: 10.3389/fimmu.2023.1131146] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
During mucosal injury, intestinal immune cells play a crucial role in eliminating invading bacteria. However, as the excessive accumulation of immune cells promotes inflammation and delays tissue repair, it is essential to identify the mechanism that limits the infiltration of immune cells to the mucosal-luminal interface. Cholesterol sulfate (CS) is the lipid product of the sulfotransferase SULT2B1 and suppresses immune reactions by inhibiting DOCK2-mediated Rac activation. In this study, we aimed to elucidate the physiological role of CS in the intestinal tract. We found that, in the small intestine and colon, CS is predominantly produced in the epithelial cells close to the lumen. While dextran sodium sulfate (DSS)-induced colitis was exacerbated in Sult2b1-deficient mice with increased prevalence of neutrophils, the elimination of either neutrophils or intestinal bacteria in Sult2b1-deficient mice attenuated disease development. Similar results were obtained when the Dock2 was genetically deleted in Sult2b1-deficient mice. In addition, we also show that indomethacin-induced ulcer formation in the small intestine was exacerbated in Sult2b1-deficient mice and was ameliorated by CS administration. Thus, our results uncover that CS acts on inflammatory neutrophils, and prevents excessive gut inflammation by inhibiting the Rac activator DOCK2. The administration of CS may be a novel therapeutic strategy for inflammatory bowel disease and non-steroidal anti-inflammatory drug-induced ulcers.
Collapse
Affiliation(s)
- Kenji Morino
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- *Correspondence: Kazufumi Kunimura, ; Yoshinori Fukui,
| | - Yuki Sugiura
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keisuke Matsubara
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Sayaka Akiyoshi
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Rae Maeda
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenichiro Hirotani
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Daiji Sakata
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- *Correspondence: Kazufumi Kunimura, ; Yoshinori Fukui,
| |
Collapse
|
22
|
Turki T, Taguchi YH. A new machine learning based computational framework identifies therapeutic targets and unveils influential genes in pancreatic islet cells. Gene 2023; 853:147038. [PMID: 36503891 DOI: 10.1016/j.gene.2022.147038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 11/04/2022] [Indexed: 11/29/2022]
Abstract
Pancreatic islets comprise a group of cells that produce hormones regulating blood glucose levels. Particularly, the alpha and beta islet cells produce glucagon and insulin to stabilize blood glucose. When beta islet cells are dysfunctional, insulin is not secreted, inducing a glucose metabolic disorder. Identifying effective therapeutic targets against the disease is a complicated task and is not yet conclusive. To close the wide gap between understanding the molecular mechanism of pancreatic islet cells and providing effective therapeutic targets, we present a computational framework to identify potential therapeutic targets against pancreatic disorders. First, we downloaded three transcriptome expression profiling datasets pertaining to pancreatic islet cells (GSE87375, GSE79457, GSE110154) from the Gene Expression Omnibus database. For each dataset, we extracted expression profiles for two cell types. We then provided these expression profiles along with the cell types to our proposed constrained optimization problem of a support vector machine and to other existing methods, selecting important genes from the expression profiles. Finally, we performed (1) an evaluation from a classification perspective which showed the superiority of our methods against the baseline; and (2) an enrichment analysis which indicated that our methods achieved better outcomes. Results for the three datasets included 44 unique genes and 10 unique transcription factors (SP1, HDAC1, EGR1, E2F1, AR, STAT6, RELA, SP3, NFKB1, and ESR1) which are reportedly related to pancreatic islet functions, diseases, and therapeutic targets.
Collapse
Affiliation(s)
- Turki Turki
- King Abdulaziz University, Department of Computer Science, Jeddah 21589, Saudi Arabia.
| | - Y-H Taguchi
- Chuo University, Department of Physics, Tokyo 112-8551, Japan.
| |
Collapse
|
23
|
Human glycoprotein-2 expressed in Brunner glands - A putative autoimmune target and link between Crohn's and coeliac disease. Clin Immunol 2023; 247:109214. [PMID: 36608744 DOI: 10.1016/j.clim.2022.109214] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
Glycoprotein 2 (GP2) is an autoantigen in Crohn's (CD) and coeliac disease (CeD). We assessed GP2-isoform (GP21-4)-expression in intestinal biopsies of paediatric patients with CD, CeD, ulcerative colitis (UC), and healthy children (HC). Transcription of GP21-4 was elevated in proximal small intestine in CeD and CD patients (only GP22/4) compared to jejunum (CeD/CD) and large bowel (CD). CeD patients demonstrated higher duodenal GP22/4-mRNA levels compared to HC/UC patients whereas CD patients showed higher GP24-mRNA levels compared to UC patients. Duodenal synthesis of only small GP2 isoforms (GP23/4) was demonstrated in epithelial cells in patients/HC and in Brunner glands (also large isoforms) with a more frequent apical location in CD/CeD patients. All four GP2 isoforms interacted with gliadin and phosphopeptidomannan. Gliadin digestion improved binding to GP2 isoforms. GP21-4 binding to CeD/CD-related antigens, elevated duodenal GP21-4-mRNA transcription, and GP2-protein secretion in Brunner glands of CeD/CD patients suggest an autoimmune CeD/CD link.
Collapse
|
24
|
Li D, Xie T, Guo T, Hu Z, Li M, Tang Y, Wu Q, Luo F, Lin Q, Wang H. Sialic acid exerts anti-inflammatory effect through inhibiting MAPK-NF-κB/AP-1 pathway and apoptosis in ulcerative colitis. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
25
|
Qu Q, Li SP, Dong Q, Du HL, Wang ZH, Ma YM, Gong XP, Ding YQ, Zhou J, Chen JY, Liu MJ, Lv WJ, Guo SN. Transcriptome profiling Revealed the potential mechanisms of Shen Lin Bai Zhu San n-butanol extract on DSS induced Colitis in Mice and LC-MS analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154645. [PMID: 36634382 DOI: 10.1016/j.phymed.2023.154645] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/24/2022] [Accepted: 01/01/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic and recurrent inflammatory disorder in gastrointestinal tract. Shen Ling Bai Zhu San (SLBZS), which has a long history of use in Traditional Chinese Medicine (TCM), has been widely used to treat gastrointestinal diseases. The isolated fractions of TCM have also been proved to possess an important potential for treating diseases, which are due to their effective components. PURPOSE In this study, we examined the possibility that SLBZS and its isolated active fractions may prevent DSS-induced colitis, and investigated the potential mechanisms by regulating genetic profile of colon. METHODS Colitis mice were induced by 2.5% DSS for 7 days, and then SLBZS and different SLBZS extracts were administrated to protect the mice for 7 days. Body weight, diarrhea, bleeding in stool, colon length, spleen weight, cytokines of serum and colon and pathology of colon were assessed. The level of Ginsenoside Rg1, Re and Rb1 in different SLBZS extracts and qualitative analysis of n-butanol extract of SLBZS (S-Nb) was performed by HPLC and LC-MS, respectively. And the effects of S-Nb on the transcriptome in colitis were investigated. RESULTS Our results showed that SLBZS and S-Nb significantly regained body weight, reduced DAI, splenomegaly and the length of colon and attenuated histological damage of the colon. Meanwhile, SLBZS and S-Nb markedly reduced the levels of TNF-α, IL-1β and IL-6 and increased the level of IL-10 in serum and colon. These effects may be associated with the high levels of Ginsenoside Rg1, Re and Rb1 and rich variety of compounds in S-Nb including 6 ginsenosides, glycyrrhizin, L-tryptophan, and so on. Transcriptome analysis revealed that S-Nb selectively regulated 103 differentially expressed genes (DEGs), 36 of which were changed in DSS-induced mice. And the genes of Per2, Per3, Npy and Serpina3m were closely related to colitis and also restored by S-Nb with different extent. Remarkably, these DEGs modulated the biological functions of colitis mice, including extracellular region, response to external stimulus, MAPK signaling pathway and arginine and proline metabolism. CONCLUSIONS These data indicated that SLBZS and S-Nb blunted DSS-induced colitis by modulating differentially expression gene profile and biological functions based on their ginsenosides and rich compounds.
Collapse
Affiliation(s)
- Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Shu-Peng Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Qi Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Hong-Liang Du
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhi-Hua Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yi-Mu Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiao-Pei Gong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yi-Qing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jing Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jia-Yan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Meng-Jie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Wei-Jie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| | - Shi-Ning Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Research Center for Veterinary Traditional Chinese Medicine and Natural Medicine Engineering Technology, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
26
|
Pirini F, Cortesi M, Tumedei MM, Zanoni M, Ravaioli S, Bravaccini S. Tumor resident microbiota and response to therapies: An insight on tissue bacterial microbiota. Front Cell Dev Biol 2023; 10:1048360. [PMID: 36684442 PMCID: PMC9845623 DOI: 10.3389/fcell.2022.1048360] [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: 09/19/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
The role of the intestinal microbiota in the promotion, progression, and response to therapies is gaining importance, but recent studies confirm the presence of microbiota also in the tumor, thus becoming a component of the tumor microenvironment. There is not much knowledge on the characteristics and mechanisms of action of the tumor resident microbiota, but there are already indications of its involvement in conditioning the response to therapies. In this review, we discuss recent publications on the interaction between microbiota and anticancer treatments, mechanisms of resistance and possible strategies for manipulating the microbiota that could improve treatments in a personalized medicine perspective.
Collapse
|
27
|
Iwanaga T, Kimura S. GP2-expressing cells: a new guardian with divergent functions in the intestine, eyes, and nose. Biomed Res 2023; 44:233-243. [PMID: 38008422 DOI: 10.2220/biomedres.44.233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
GP (glycoprotein)-2, originally identified as a predominant membranous component of pancreatic acinar cells, has attracted the interest of researchers in mucosal immunology for its role as a functional molecule specific for antigen-sampling cells in the intestinal Peyer's patches. GP2 is involved in the detection of pathological bacteria and is also histologically useful for the identification of the M cell lineage and their differentiation in lymphoid tissues. Subsequent immunohistochemistry for GP2 has revealed a broad distribution of M cells and related cells in the nasopharyngeal lymphoid tissues, conjunctiva, tear duct, and airway. Especially, GP2 cells in the paranasal sinuses and tear duct have been identified as novel types of epithelial cells. The systematic administration of RANKL can induce extra-M cells in conventional epithelia of body. The production and release of GP2 by conjunctival goblet cells and several mucous glands suggests leading roles for mucous cells in protection, including the entrapment of microorganisms for infections. The ocular surface and conjunctiva are connected to the lacrimal sac, nasolacrimal duct, and further nasal cavity, comprising another canal that passes through the body. The broad distribution of GP2-expressingcells may indicate its function as a new guardian in the intestine, eyes, and nose, all of which are exposed to external milieu.
Collapse
Affiliation(s)
- Toshihiko Iwanaga
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Shunsuke Kimura
- Division of Biochemis- try, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| |
Collapse
|
28
|
Shibata K, Motozono C, Nagae M, Shimizu T, Ishikawa E, Motooka D, Okuzaki D, Izumi Y, Takahashi M, Fujimori N, Wing JB, Hayano T, Asai Y, Bamba T, Ogawa Y, Furutani-Seiki M, Shirai M, Yamasaki S. Symbiotic bacteria-dependent expansion of MR1-reactive T cells causes autoimmunity in the absence of Bcl11b. Nat Commun 2022; 13:6948. [PMID: 36376329 PMCID: PMC9663695 DOI: 10.1038/s41467-022-34802-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
MHC class I-related protein 1 (MR1) is a metabolite-presenting molecule that restricts MR1-reactive T cells including mucosal-associated invariant T (MAIT) cells. In contrast to MAIT cells, the function of other MR1-restricted T cell subsets is largely unknown. Here, we report that mice in which a T cell-specific transcription factor, B-cell lymphoma/leukemia 11B (Bcl11b), was ablated in immature thymocytes (Bcl11b∆iThy mice) develop chronic inflammation. Bcl11b∆iThy mice lack conventional T cells and MAIT cells, whereas CD4+IL-18R+ αβ T cells expressing skewed Traj33 (Jα33)+ T cell receptors (TCR) accumulate in the periphery, which are necessary and sufficient for the pathogenesis. The disorders observed in Bcl11b∆iThy mice are ameliorated by MR1-deficiency, transfer of conventional T cells, or germ-free conditions. We further show the crystal structure of the TCR expressed by Traj33+ T cells expanded in Bcl11b∆iThy mice. Overall, we establish that MR1-reactive T cells have pathogenic potential.
Collapse
Affiliation(s)
- Kensuke Shibata
- grid.268397.10000 0001 0660 7960Department of Microbiology and Immunology, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan ,grid.177174.30000 0001 2242 4849Department of Ophthalmology, Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan ,grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Chihiro Motozono
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.274841.c0000 0001 0660 6749Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0871 Japan
| | - Masamichi Nagae
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Takashi Shimizu
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Eri Ishikawa
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Daisuke Motooka
- grid.136593.b0000 0004 0373 3971Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Daisuke Okuzaki
- grid.136593.b0000 0004 0373 3971Single Cell Genomics, Human Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Yoshihiro Izumi
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Masatomo Takahashi
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Nao Fujimori
- grid.177174.30000 0001 2242 4849Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - James B. Wing
- grid.136593.b0000 0004 0373 3971Laboratory of Human Immunology (Single Cell Immunology), World Premier International Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Takahide Hayano
- grid.268397.10000 0001 0660 7960Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Yoshiyuki Asai
- grid.268397.10000 0001 0660 7960Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Takeshi Bamba
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Yoshihiro Ogawa
- grid.177174.30000 0001 2242 4849Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan ,grid.419082.60000 0004 1754 9200Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology, Tokyo, 100-0004 Japan ,grid.27476.300000 0001 0943 978XDepartment of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601 Japan
| | - Makoto Furutani-Seiki
- grid.268397.10000 0001 0660 7960Systems Biochemistry in Pathology and Regeneration, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Mutsunori Shirai
- grid.268397.10000 0001 0660 7960Department of Microbiology and Immunology, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Sho Yamasaki
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.177174.30000 0001 2242 4849Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan ,grid.136304.30000 0004 0370 1101Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, 260-8673 Japan
| |
Collapse
|
29
|
Luda KM, Da Silva C, Ahmadi F, Mowat AM, Ohno H, Kotarsky K, Agace WW. Identification and characterization of murine glycoprotein 2-expressing intestinal dendritic cells. Scand J Immunol 2022; 96:e13219. [PMID: 37807915 PMCID: PMC9786990 DOI: 10.1111/sji.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
The intestinal lamina propria (LP) contains distinct subsets of classical dendritic cells (cDC), each playing key non-redundant roles in intestinal immune homeostasis. Here, we show that glycoprotein 2 (GP2), a GPI-anchored protein and receptor for bacterial type-I fimbriae, is selectively expressed by CD103+CD11b+ cDC in the murine small intestine (SI). GP2 expression was induced on CD103+CD11b+ cDC within the SI-LP and was regulated by IRF4, TGFβR1- and retinoic acid signalling. Mice selectively lacking Gp2 on CD103+CD11b+ cDC (huLang-Cre.gp2fl/fl mice) had normal numbers and proportions of innate and adaptive immune cells in the SI-LP suggesting that GP2 expression by CD103+CD11b+ cDC is not required for intestinal immune homoeostasis.
Collapse
Affiliation(s)
- Katarzyna M Luda
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Clement Da Silva
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fatemeh Ahmadi
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Allan Mcl Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow, UK
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Knut Kotarsky
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - William W Agace
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Mucosal Immunology Laboratory, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
30
|
Uhlig R, Günther K, Bröker N, Gorbokon N, Lennartz M, Dwertmann Rico S, Reiswich V, Viehweger F, Büscheck F, Kluth M, Hube-Magg C, Hinsch A, Fraune C, Bernreuther C, Lebok P, Sauter G, Izbicki JR, Steurer S, Burandt E, Marx AH, Krech T, Simon R, Minner S, Clauditz TS, Jacobsen F. Diagnostic and prognostic role of pancreatic secretory granule membrane major glycoprotein 2 (GP2) immunohistochemistry: A TMA study on 27,681 tumors. Pathol Res Pract 2022; 238:154123. [PMID: 36137400 DOI: 10.1016/j.prp.2022.154123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022]
Abstract
Pancreatic secretory granule membrane major glycoprotein 2 (GP2) is a membrane component of zymogen granules which is abundantly secreted by pancreatic acinar cells. Because RNA based analyses suggest a strict limitation of GP2 expression to the pancreas in normal tissues, and a strong preference to pancreatic cancer among tumors, GP2 expression analysis might have diagnostic utility. To better understand the role of GP2 protein expression, GP2 was successfully analyzed in 27,965 tumor samples from 132 different tumor types and subtypes as well as 8 samples each of 76 different normal tissue types by immunohistochemistry in a tissue microarray format (TMA). GP2 immunostaining was seen in 14 of 16 (87.5 %) acinar cell carcinomas, 6 of 507 (1.2 %) ductal adenocarcinomas, and 3 of 99 neuroendocrine neoplasms of the pancreas (3.0 %). GP2 was also found in 23 extra-pancreatic tumor entities including several types of neuroendocrine neoplasms (14.3-58.8 %), prostatic adenocarcinomas (8.2-18.8 %), various other adenocarcinomas (0.1-7.7 %), and several categories of benign and malignant salivary gland tumors (2.3-3.1 %). A strong GP2 positivity was only seen in 6 tumor categories including 50 % of 16 pancreatic acinus cell carcinomas, 11.8 % of 17 neuroendocrine tumors of the lung, 1.3 % of 80 primary Gleason 4 + 4 % and 0.6 % of 181 recurrent prostate cancers, as well as 0.8 % of 133 adenocarcinomas of the lung. In a cohort of 14,747 prostate cancers with follow up data, GP2 immunostaining was strongly linked to advanced pT stage, high Gleason grade, lymph node metastasis, and recurrence free survival (p < 0.0001 each). The prognostic impact of GP2 positivity was independent of established parameters in TMPRSS2:ERG fusion-negative cancers (p < 0.0001). In summary, our data show that GP2 is preferentially expressed in acinar cell carcinomas of the pancreas but the glycoprotein can - rarely - also be expressed in a variety of other tumor entities.
Collapse
Affiliation(s)
- Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karin Günther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Bröker
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R Izbicki
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
31
|
Surfactin Mitigates a High-Fat Diet and Streptozotocin-Induced Type 2 Diabetes through Improving Pancreatic Dysfunction and Inhibiting Inflammatory Response. Int J Mol Sci 2022; 23:ijms231911086. [PMID: 36232419 PMCID: PMC9570334 DOI: 10.3390/ijms231911086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
Surfactin from Bacillus amyloliquefaciens fmb50 was utilized to treat mice with type 2 diabetes (T2DM) induced by a high-fat diet/streptozotocin (HFD/STZ). Our group’s earlier research indicated that surfactin could lower blood glucose and mitigate liver dysfunction to further improve HFD/STZ-induced T2DM through modulating intestinal microbiota. Thus, we further investigated the effects of surfactin on the pancreas and colon in mice with T2DM to elucidate the detailed mechanism. In the present study, mice with HFD/STZ-induced T2DM had their pancreatic and colon inflammation, oxidative stress, and endoplasmic reticulum stress (ERS) reduced when given oral surfactin at a dose of 80 mg/kg body weight. According to further research, surfactin also improved glucose metabolism by activating the phosphatidylinositol kinase (PI3K)/protein kinase B (Akt) signaling pathway, further protecting islets β-cell, promoting insulin secretion, inhibiting glucagon release and mitigating pancreas dysfunction. Additionally, after surfactin treatment, the colon levels of the tight junction proteins Occludin and Claudin-1 of T2DM mice were considerably increased by 130.64% and by 36.40%, respectively. These findings revealed that surfactin not only ameliorated HFD/STZ-induced pancreas inflammation and dysfunction and preserved intestinal barrier dysfunction and gut microbiota homeostasis but also enhanced insulin sensitivity and glucose homeostasis in T2DM mice. Finally, in the further experiment, we were able to demonstrate that early surfactin intervention might delay the development of T2DM caused by HFD/STZ, according to critical biochemical parameters in serum.
Collapse
|
32
|
Pathological features-based targeted delivery strategies in IBD therapy: A mini review. Biomed Pharmacother 2022; 151:113079. [PMID: 35605297 DOI: 10.1016/j.biopha.2022.113079] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is characterized by a complex and dysfunctional immune response. Currently, IBD is incurable, and patients with IBD often need to take drugs for life. However, as the traditional systemic treatment strategies for IBD do not target the site of inflammation, only limited efficacy can be obtained from them. Moreover, the possibility of serious side effects stemming from the systemic administration or redistribution of drugs in the body is high when conventional drug formulations are used. Therefore, a targeted drug-delivery system for IBD should be considered. Based on the pathological features related to IBD, the new targeted drug-delivery strategy can directly transfer the drug to the inflammatory site, thus enhancing the accumulation of the drugs and reducing side effects. This article reviews the pathological features of IBD and the application of the IBD-targeted delivery system based on different pathological features, and discusses the challenges and new prospects in this field.
Collapse
|
33
|
Gut microbiota alternation under the intestinal epithelium-specific knockout of mouse Piga gene. Sci Rep 2022; 12:10812. [PMID: 35752737 PMCID: PMC9233684 DOI: 10.1038/s41598-022-15150-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/22/2022] [Indexed: 11/08/2022] Open
Abstract
Crosstalk between the gut microbiota and intestinal epithelium shapes the gut environment and profoundly influences the intestinal immune homeostasis. Glycosylphosphatidylinositol anchored proteins (GPI – APs) contribute to a variety of gut-associated immune functions, including microbial surveillance and defense, and epithelial cell polarity. Properly polarised epithelial cells are essential for the establishment of the barrier function of gut epithelia. The Piga gene is one among seven genes that encode for an enzyme which is involved in the first step of GPI-anchor biosynthesis. This is the first study reporting a knockout of the intestinal epithelial cell-specific Piga gene (Piga-/-) and its association with the gut microbiota in mice using a whole metagenome shotgun-based sequencing approach. An overall reduced microbiota diversity has been observed in the Piga-/- group as compared to the control group (ANOVA p = 0.34). The taxonomic biomarkers, namely: Gammaproteobacteria (class), Enterobacterales (order), Enterobacteriaceae (family), Escherichia (genus), Proteus (genus) and Escherichia coli (species), increased more in the Piga-/- mice as compared to in the control group. Further, the pathogenic E. coli strains, namely E. coli O157:H7 str. EDL 933 (EHEC), E. coli CFT073 (UPEC) and E. coli 536 (UPEC), were found in the Piga-/- mice which also harbored virulence factor transporters. In addition, the taxa responsible for short chain fatty acid production were decreased in the Piga-/- group. The Piga-/- mice gut harbored an increased number of microbial functions responsible for the survival of pathogens in the inflamed gut environment. Our observations clearly indicate that the Piga-/- mice gut might have an overall enhancement in pathogenic behaviour and reduced capabilities beneficial to health.
Collapse
|
34
|
Martinez-Ramirez AS, Borders TL, Paul L, Schipma M, Wang X, Korobova F, Wright CV, Sosa-Pineda B. Specific Temporal Requirement of Prox1 Activity During Pancreatic Acinar Cell Development. GASTRO HEP ADVANCES 2022; 1:807-823. [PMID: 37829188 PMCID: PMC10569262 DOI: 10.1016/j.gastha.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
BACKGROUND AND AIMS An interactive regulatory network assembled through the induction and downregulation of distinct transcription factors governs acinar cell maturation. Understanding how this network is built is relevant for protocols of directed pancreatic acinar differentiation. The murine transcription factor Prox1 is highly expressed in multipotent pancreatic progenitors and in various mature pancreatic cell types except for acinar cells. In this study, we investigated when is Prox1 expression terminated in developing acinar cells and the potential involvement of its activity in acinar cell specification/differentiation. We also investigated the effects of sustained Prox1 expression in acinar maturation and maintenance. METHODS Prox1 acinar expression was analyzed by immunofluorescence and confocal microscopy. Prox1-null embryos (Prox1GFPCre/Δ), Prox1AcOE transgenic mice, histologic and immunostaining methods, transmission electron microscopy, functional assays, and quantitative RNA and RNA-sequencing methods were used to investigate the effects of Prox1 functional deficiency and sustained Prox1 expression in acinar maturation and homeostasis. RESULTS Immunostaining results reveal transient Prox1 expression in newly committed embryonic acinar cells. RNA-sequencing demonstrate precocious expression of multiple "late" acinar genes in the pancreas of Prox1GFPCre/Δ embryos. Prox1AcOE transgenic mice carrying sustained Prox1 acinar expression have relatively normal pancreas development. In contrast, Prox1AcOE adult mice have severe pancreatic alterations involving reduced acinar gene expression, abnormal acinar secretory granules, acinar atrophy, increased endoplasmic reticulum stress, and mild chronic inflammation. CONCLUSION Prox1 transient expression in early acinar cells is necessary for correct sequential gene expression. Prox1 expression is terminated in developing acinar cells to complete maturation and to preserve homeostasis.
Collapse
Affiliation(s)
- Angelica S. Martinez-Ramirez
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Thomas L. Borders
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Leena Paul
- Department of Genetics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Matthew Schipma
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Xinkun Wang
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Farida Korobova
- Center for Advanced Microscopy, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Christopher V. Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Beatriz Sosa-Pineda
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
35
|
Zhang Z, Tanaka I, Pan Z, Ernst PB, Kiyono H, Kurashima Y. Intestinal homeostasis and inflammation: gut microbiota at the crossroads of pancreas-intestinal barrier axis. Eur J Immunol 2022; 52:1035-1046. [PMID: 35476255 PMCID: PMC9540119 DOI: 10.1002/eji.202149532] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/24/2022]
Abstract
The pancreas contains exocrine glands, which release enzymes (e.g., amylase, trypsin, and lipase) that are important for digestion and islets, which produce hormones. Digestive enzymes and hormones are secreted from the pancreas into the duodenum and bloodstream, respectively. Growing evidence suggests that the roles of the pancreas extend to not only the secretion of digestive enzymes and hormones but also to the regulation of intestinal homeostasis and inflammation (e.g., mucosal defense to pathogens and pathobionts). Organ crosstalk between the pancreas and intestine is linked to a range of physiological, immunological, and pathological activities, such as the regulation of the gut microbiota by the pancreatic proteins and lipids, the retroaction of the gut microbiota on the pancreas, the relationship between inflammatory bowel disease, and pancreatic diseases. We herein discuss the current understanding of the pancreas–intestinal barrier axis and the control of commensal bacteria in intestinal inflammation.
Collapse
Affiliation(s)
- Zhongwei Zhang
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Izumi Tanaka
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Zhen Pan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Peter B Ernst
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, San Diego, CA, 92093-0956, USA.,Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA, 92093-0956, USA.,Departments of Medicine and Pathology, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA, 92093-0956, USA.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Hiroshi Kiyono
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, San Diego, CA, 92093-0956, USA.,Departments of Medicine and Pathology, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA, 92093-0956, USA.,Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Human Mucosal Vaccinology, Chiba University, Chiba, 260-8670, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.,Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, San Diego, CA, 92093-0956, USA.,Departments of Medicine and Pathology, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA, 92093-0956, USA.,Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Human Mucosal Vaccinology, Chiba University, Chiba, 260-8670, Japan.,Institute for Advanced Academic Research, Chiba University, Chiba, 260-8670, Japan
| |
Collapse
|
36
|
Stsiapanava A, Xu C, Nishio S, Han L, Yamakawa N, Carroni M, Tunyasuvunakool K, Jumper J, de Sanctis D, Wu B, Jovine L. Structure of the decoy module of human glycoprotein 2 and uromodulin and its interaction with bacterial adhesin FimH. Nat Struct Mol Biol 2022; 29:190-193. [PMID: 35273390 PMCID: PMC8930769 DOI: 10.1038/s41594-022-00729-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022]
Abstract
Glycoprotein 2 (GP2) and uromodulin (UMOD) filaments protect against gastrointestinal and urinary tract infections by acting as decoys for bacterial fimbrial lectin FimH. By combining AlphaFold2 predictions with X-ray crystallography and cryo-EM, we show that these proteins contain a bipartite decoy module whose new fold presents the high-mannose glycan recognized by FimH. The structure rationalizes UMOD mutations associated with kidney diseases and visualizes a key epitope implicated in cast nephropathy. AlphaFold2 predictions, X-ray crystallography and cryo-EM analyses reveal how related human glycoproteins GP2 and uromodulin catch pathogenic bacteria by presenting a high-mannose glycan that acts as a decoy for fimbrial adhesin FimH.
Collapse
Affiliation(s)
- Alena Stsiapanava
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Chenrui Xu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,NTU Institute of Structural Biology, Nanyang Technological University, Singapore, Singapore
| | - Shunsuke Nishio
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Ling Han
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Nao Yamakawa
- US 41-UMS 2014-PLBS, Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Marta Carroni
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | | | | | | | - Bin Wu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,NTU Institute of Structural Biology, Nanyang Technological University, Singapore, Singapore
| | - Luca Jovine
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
37
|
[The role of the microbiome in diseases of the pancreas]. Internist (Berl) 2022; 63:372-378. [PMID: 35175369 DOI: 10.1007/s00108-022-01276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The human body is inhabited by diverse microorganisms. Together, this so-called microbiome exerts important metabolic functions and contributes to the maintenance of health. At the same time, shifts in the microbiome composition may lead to disease. OBJECTIVES Review of the current literature about the role of the microbiome in diseases of the pancreas. MATERIALS AND METHODS Literature search in PubMed and Embase. RESULTS The exocrine pancreas is a major factor determining the composition and stability of the intestinal microbiome even in healthy people without pancreatic disease. Inflammatory diseases of the pancreas such as acute or chronic pancreatitis lead to reduced microbial diversity, loss of gut barrier stabilizing bacteria and an increase in facultative pathogens like Escherichia or Enterococcus. Even pancreatic cancer tissue harbours microbiota and mice models have shown that the growth of pancreatic cancer can be inhibited by microbiota ablation. CONCLUSIONS Inflammatory diseases of the pancreas lead to gut microbiome dysbiosis and tumor microbiota probably play a role in the development of pancreatic cancer. Until now, however, there is no proof that therapeutic microbiota modulation in individuals with pancreatic disease can improve mortality or quality of life. At this point, the analysis of the microbiome in pancreatic disease should only be performed in scientific studies.
Collapse
|
38
|
Adhesion of enteropathogenic, enterotoxigenic and commensal Escherichia coli to the Major Zymogen Granule Membrane Glycoprotein 2. Appl Environ Microbiol 2022; 88:e0227921. [PMID: 35020452 PMCID: PMC8904060 DOI: 10.1128/aem.02279-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pathogenic bacteria, such as enteropathogenic Escherichia coli (EPEC) and enterotoxigenic E. coli (ETEC), cause diarrhea in mammals. In particular, E. coli colonizes and infects the gastrointestinal tract via type 1 fimbriae (T1F). Here, the major zymogen granule membrane glycoprotein 2 (GP2) acts as a host cell receptor. GP2 is also secreted by the pancreas and various mucous glands, interacting with luminal type 1 fimbriae-positive E. coli. It is unknown whether GP2 isoforms demonstrate specific E. coli pathotype binding. In this study, we investigated interactions of human, porcine, and bovine EPEC and ETEC, as well as commensal E. coli isolates with human, porcine, and bovine GP2. We first defined pathotype- and host-associated FimH variants. Second, we could prove that GP2 isoforms bound to FimH variants to various degrees. However, the GP2-FimH interactions did not seem to be influenced by the host specificity of E. coli. In contrast, soluble GP2 affected ETEC infection and phagocytosis rates of macrophages. Preincubation of the ETEC pathotype with GP2 reduced the infection of cell lines. Furthermore, preincubation of E. coli with GP2 improved the phagocytosis rate of macrophages. Our findings suggest that GP2 plays a role in the defense against E. coli infection and in the corresponding host immune response. IMPORTANCE Infection by pathogenic bacteria, such as certain Escherichia coli pathotypes, results in diarrhea in mammals. Pathogens, including zoonotic agents, can infect different hosts or show host specificity. There are Escherichia coli strains which are frequently transmitted between humans and animals, whereas other Escherichia coli strains tend to colonize only one host. This host specificity is still not fully understood. We show that glycoprotein 2 is a selective receptor for particular Escherichia coli strains or variants of the adhesin FimH but not a selector for a species-specific Escherichia coli group. We demonstrate that GP2 is involved in the regulation of colonization and infection and thus represents a molecule of interest for the prevention or treatment of disease.
Collapse
|
39
|
Minamisawa M, Sato Y, Ishiguro E, Taniai T, Sakamoto T, Kawai G, Saito T, Saido TC. Amelioration of Alzheimer's Disease by Gut-Pancreas-Liver-Brain Interaction in an App Knock-In Mouse Model. Life (Basel) 2021; 12:34. [PMID: 35054427 PMCID: PMC8778338 DOI: 10.3390/life12010034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
In this study, we observed disease progression, changes in the gut microbiota, and interactions among the brain, liver, pancreas, and intestine in a mouse model of Alzheimer's disease (AD), in addition to attempting to inhibit disease progression through the dietary supplementation of L-arginine and limonoids. Wild-type mice (WC) and AD mice were fed a normal diet (AC), a diet supplemented with L-arginine and limonoids (ALA), or a diet containing only limonoids (AL) for 12-64 weeks. The normal diet-fed WC and AC mice showed a decrease in the diversity of the gut microbiota, with an increase in the Firmicutes/Bacteroidetes ratio, and bacterial translocation. Considerable bacterial translocation to the pancreas and intense inflammation of the pancreas, liver, brain, and intestinal tissues were observed in the AC mice from alterations in the gut microbiota. The ALA diet or AL diet-fed mice showed increased diversity of the bacterial flora and suppressed oxidative stress and inflammatory responses in hepatocytes and pancreatic cells, bacterial translocation, and neurodegeneration of the brain. These findings suggest that L-arginine and limonoids help in maintaining the homeostasis of the gut microbiota, pancreas, liver, brain, and gut in AD mice.
Collapse
Affiliation(s)
- Mayumi Minamisawa
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
- Education Center, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba 275-0023, Japan;
| | - Yuma Sato
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
| | | | - Tetsuyuki Taniai
- Education Center, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba 275-0023, Japan;
| | - Taiichi Sakamoto
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
| | - Gota Kawai
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
| | - Takashi Saito
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, Saitama 351-0198, Japan; (T.S.); (T.C.S.)
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Takaomi C. Saido
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, Saitama 351-0198, Japan; (T.S.); (T.C.S.)
| |
Collapse
|
40
|
Lin Y, Nakatochi M, Sasahira N, Ueno M, Egawa N, Adachi Y, Kikuchi S. Glycoprotein 2 in health and disease: lifting the veil. Genes Environ 2021; 43:53. [PMID: 34861888 PMCID: PMC8641183 DOI: 10.1186/s41021-021-00229-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/16/2021] [Indexed: 11/10/2022] Open
Abstract
In 2020, we discovered glycoprotein 2 (GP2) variants associated with pancreatic cancer susceptibility in a genome-wide association study involving the Japanese population. Individuals carrying a missense coding variant (rs78193826) in the GP2 gene resulting in a p.V432M substitution had an approximately 1.5-fold higher risk of developing pancreatic cancer than those without this variant. GP2 is expressed on the inner surface of zymogen granules in pancreatic acinar cells, which are responsible for the sorting, storage and secretion of digestive enzymes. Upon neuronal, hormonal, or other stimulation, GP2 is cleaved from the membrane of zymogen granules and then secreted into the pancreatic duct and intestinal lumen. While the functions of GP2 remain poorly understood, emerging evidence suggests that it plays an antibacterial role in the gastrointestinal tract after being secreted from pancreatic acinar cells. Impaired GP2 functions may facilitate the adhesion of bacteria to the intestinal mucosa. In this review article, we summarize the role of GP2 in health and disease, emphasizing its functions in the gastrointestinal tract, as well as genetic variations in the GP2 gene and their associations with disease susceptibility. We hope that its robust genetic associations with pancreatic cancer, coupled with its emerging role in gastrointestinal mucosal immunity, will spur renewed research interest in GP2, which has been understudied over the past 30 years compared with its paralog uromodulin (UMOD).
Collapse
Affiliation(s)
- Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, 480-1195, Nagakute, Aichi, Japan.
| | - Masahiro Nakatochi
- Division of Public Health Informatics, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, 461-8673, Nagoya, Japan
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 135-8550, Tokyo, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, 241-8515, Yokohama, Japan
| | - Naoto Egawa
- Department of Internal Medicine, Tokyo Metropolitan Matsuzawa Hospital, 156- 0057, Tokyo, Japan
| | - Yasushi Adachi
- Division of Gastroenterology, Department of Internal Medicine, Sapporo Shirakaba- dai Hospital, 062-0052, Sapporo, Japan
| | - Shogo Kikuchi
- Department of Public Health, Aichi Medical University School of Medicine, 480-1195, Nagakute, Aichi, Japan
| |
Collapse
|
41
|
Liu H, Liang J, Zhong Y, Xiao G, Efferth T, Georgiev MI, Vargas-De-La-Cruz C, Bajpai VK, Caprioli G, Liu J, Lin J, Wu H, Peng L, Li Y, Ma L, Xiao J, Wang Q. Dendrobium officinale Polysaccharide Alleviates Intestinal Inflammation by Promoting Small Extracellular Vesicle Packaging of miR-433-3p. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13510-13523. [PMID: 34739249 DOI: 10.1021/acs.jafc.1c05134] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dendrobium officinale polysaccharide (DOP) attenuates inflammatory bowel disease (IBD), but its role in regulating cross-talk between intestinal epithelial cells (IEC) and macrophages against IBD is unclear. This study aimed to investigate DOP protective effects on the intestinal inflammatory response through regulation by miRNA in small extracellular vesicles (sEVs). Our results show that DOP interfered with the secretion of small extracellular vesicles (DIEs) by IEC, which reduced the levels of inflammatory mediators. Increased miR-433-3p expression in DIEs was identified as an important protector against intestinal inflammation. DOP regulated the loading of miR-433-3p by hnRNPA2B1 into the intestinal sEV to increase the abundance of miR-433-3p. DIEs delivered miR-433-3p to lipopolysaccharide-induced macrophages and targeted the MAPK8 gene, leading to inhibition of the MAPK signaling pathway and reduced production of inflammatory cytokines. One protective mechanism of DOP is mediated by intestinal sEV containing miR-433-3p, which is a potential therapeutic agent for the prevention of inflammatory factor accumulation from excessive intestinal macrophage activity and for restoring homeostasis in the intestinal microenvironment.
Collapse
Affiliation(s)
- Huifan Liu
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou City, Guangdong 510642, China.,College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Jiaxi Liang
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou City, Guangdong 510642, China.,College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Yuming Zhong
- College of Resources and Environment, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Gengsheng Xiao
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou City, Guangdong 510642, China.,College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Milen I Georgiev
- University of Agronomic Science and Veterinary Medicine, 59 Marasti Blvd, 011464 Bucharest, Romania.,Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Boulevard, 4000 Plovdiv, Bulgaria
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Enseñanza e Investigación en Bacteriología Alimentaria (CLEIBA), Universidad Nacional Mayor de San Marcos, Lima 15001, Perú.,Research Group Biotechnology and Omics in Life Sciences, Universidad Nacional Mayor de San Marcos, Lima 15001, Perú
| | - Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University, 30 Pildong-ro 1-gil, Seoul 04620, Republic of Korea
| | - Giovanni Caprioli
- School of Pharmacy, University of Camerino, Via Sant'Agostino 1, 62032 Camerino, MC, Italy
| | - Jianliang Liu
- Modern Agriculture Research Center, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Jintian Lin
- College of Agriculture and Biology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Hui Wu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Lianxin Peng
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Chengdu University, Chengdu, Sichuan 610106, China
| | - Yongjun Li
- Guangdong Jiangmen Chinese Medical College, Jiangmen City 529000, Guangdong China
| | - Lukai Ma
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou City, Guangdong 510642, China.,College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain
| | - Qin Wang
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Guangzhou City, Guangdong 510642, China.,College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| |
Collapse
|
42
|
Cucinotta U, Romano C, Dipasquale V. Pancreatic Associated Manifestations in Pediatric Inflammatory Bowel Diseases. Genes (Basel) 2021; 12:1372. [PMID: 34573354 PMCID: PMC8465218 DOI: 10.3390/genes12091372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 08/28/2021] [Accepted: 08/29/2021] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic relapsing inflammatory conditions of the gastrointestinal tract, encompassing Crohn's disease (CD), ulcerative colitis (UC) and inflammatory bowel disease unclassified (IBD-U). They are currently considered as systemic disorders determined by a set of genetic predispositions, individual susceptibility and environmental triggers, potentially able to involve other organs and systems than the gastrointestinal tract. A large number of patients experiences one or more extraintestinal manifestations (EIMs), whose sites affected are mostly represented by the joints, skin, bones, liver, eyes, and pancreas. Pancreatic abnormalities are not uncommon and are often underestimated, encompassing acute and chronic pancreatitis, autoimmune pancreatitis, exocrine pancreatic insufficiency and asymptomatic elevation of pancreatic enzymes. In most cases they are the result of environmental triggers. However, several genetic polymorphisms may play a role as precipitating factors or contributing to a more severe course. The aim of this paper is to provide an updated overview on the available evidence concerning the etiology, pathogenesis and clinical presentation of pancreatic diseases in IBD pediatric patients.
Collapse
Affiliation(s)
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood “G. Barresi”, University of Messina, 98124 Messina, Italy; (U.C.); (V.D.)
| | | |
Collapse
|
43
|
Pancreatic Disorders in Children with Inflammatory Bowel Disease. ACTA ACUST UNITED AC 2021; 57:medicina57050473. [PMID: 34064706 PMCID: PMC8151997 DOI: 10.3390/medicina57050473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/26/2021] [Accepted: 05/09/2021] [Indexed: 02/06/2023]
Abstract
Background and Objectives: Inflammatory bowel disease (IBD) is a chronic condition and mainly affects the intestines, however, the involvement of the other organs of the gastrointestinal tract (upper part, pancreas, and liver) have been observed. The coexistence of IBD with pancreatic pathology is rare, however, it has been diagnosed more frequently during recent years in the pediatric population. This article reviews the current literature on the most common pancreatic diseases associated with IBD in the pediatric population and their relationship with IBD activity and treatment. Materials and Methods: We performed a systematic review of data from published studies on pancreatic disorders, also reported as extraintestinal manifestations (EIMs), among children with IBD. We searched PubMed and Web of Science to identify eligible studies published prior to 25 April 2020. Results: Forty-four papers were chosen for analysis after a detailed inspection, which aimed to keep only the research studies (case control studies and cohort studies) or case reports on children and only those which were written in English. The manifestations of IBD-associated pancreatic disorders range from asymptomatic increase in pancreatic enzymes activity to severe disease such as acute pancreatitis. Acute pancreatitis (AP) induced by drugs, mainly thiopurine, seems to be the most- often-reported pancreatic disease associated with IBD in children. AP associated with other than drug etiologies, and chronic pancreatitis (CP), are rarely observed in the course of pediatric IBD. The pancreatic involvement can be strictly related to the activity of IBD and can also precede the diagnosis of IBD in some pediatric patients. The course of AP is mild in most cases and may occasionally lead to the development of CP, mainly in cases with a genetic predisposition. Conclusions: The involvement of the pancreas in the course of IBD may be considered as an EIM or a separate co-morbid disease, but it can also be a side effect of IBD therapy, therefore a differential diagnosis should always be performed. As the number of IBD incidences with concomitant pancreatic diseases is constantly increasing in the pediatric population, it is important to include pancreatic enzymes level measurement in the workup of IBD.
Collapse
|