1
|
Ehrencrona E, Gallego P, Trillo‐Muyo S, Garcia‐Bonete M, Recktenwald CV, Hansson GC, Johansson MEV. The structure of FCGBP is formed as a disulfide-mediated homodimer between its C-terminal domains. FEBS J 2025; 292:582-601. [PMID: 39754272 PMCID: PMC11796319 DOI: 10.1111/febs.17383] [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: 07/18/2024] [Revised: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025]
Abstract
Mucus in the colon is crucial for intestinal homeostasis by forming a barrier that separates microbes from the epithelium. This is achieved by the structural arrangement of the major mucus proteins, such as MUC2 and FCGBP, both of which are comprised of several von Willebrand D domains (vWD) and assemblies. Numerous disulfide bonds stabilise these domains, and intermolecular bonds generate multimers of MUC2. The oligomeric nature of FCGBP is not known. Human hFCGBP contains 13 vWD domains whereas mouse mFCGBP consists of only 7. We found unpaired cysteines in the vWD1 (human and mouse) and vWD5 (mouse)/vWD11 (human) assemblies which were not involved in disulfide bonds. However, the most C-terminal vWD domains, vWD7 (mouse)/vWD13 (human), formed disulfide-linked dimers. The intermolecular bond between C5284 and C5403 of human hFCGBP was observed by using mass spectrometry to generate the dimer. Cryo-EM structure analysis of recombinant mouse mFCGBP revealed a compact dimer with two symmetric intermolecular disulfide bonds between C2462 and C2581, corresponding to the dimerising cysteines in the human hFCGBP. This compact conformation involves interactions between the vWD assemblies, but although the domains involved at the interface are the same, the nature of the interactions differ. Mouse mFCGBP was also found to exist in a semi-extended conformation. These different interactions offer insights into the dynamic nature of the FCGBP homodimer.
Collapse
Affiliation(s)
- Erik Ehrencrona
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| | - Pablo Gallego
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| | - Sergio Trillo‐Muyo
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| | - Maria‐Jose Garcia‐Bonete
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| | - Christian V. Recktenwald
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| | - Malin E. V. Johansson
- Department of Medical Biochemistry and Cell BiologyInstitute of Biomedicine, University of GothenburgSweden
| |
Collapse
|
2
|
Yue N, Hu P, Tian C, Kong C, Zhao H, Zhang Y, Yao J, Wei Y, Li D, Wang L. Dissecting Innate and Adaptive Immunity in Inflammatory Bowel Disease: Immune Compartmentalization, Microbiota Crosstalk, and Emerging Therapies. J Inflamm Res 2024; 17:9987-10014. [PMID: 39634289 PMCID: PMC11615095 DOI: 10.2147/jir.s492079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
The intestinal immune system is the largest immune organ in the human body. Excessive immune response to intestinal cavity induced by harmful stimuli including pathogens, foreign substances and food antigens is an important cause of inflammatory diseases such as celiac disease and inflammatory bowel disease (IBD). Although great progress has been made in the treatment of IBD by some immune-related biotherapeutic products, yet a considerable proportion of IBD patients remain unresponsive or immune tolerant to immunotherapeutic strategy. Therefore, it is necessary to further understand the mechanism of immune cell populations involved in enteritis, including dendritic cells, macrophages and natural lymphocytes, in the steady-state immune tolerance of IBD, in order to find effective IBD therapy. In this review, we discussed the important role of innate and adaptive immunity in the development of IBD. And the relationship between intestinal immune system disorders and microflora crosstalk were also presented. We also focus on the new findings in the field of T cell immunity, which might identify novel cytokines, chemokines or anti-cytokine antibodies as new approaches for the treatment of IBD.
Collapse
Affiliation(s)
- Ningning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Peng Hu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Chengmei Tian
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Chen Kong
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Hailan Zhao
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Yuqi Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Defeng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Lisheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| |
Collapse
|
3
|
Zeng L, Zeng J, He J, Li Y, Li C, Lin Z, Chen G, Wu H, Zhou L. FCGBP functions as a tumor suppressor gene in head and neck squamous cell carcinoma. Discov Oncol 2024; 15:704. [PMID: 39580769 PMCID: PMC11586324 DOI: 10.1007/s12672-024-01607-8] [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: 06/14/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
PURPOSE The pathogenesis of head and neck squamous cell carcinoma (HNSCC) was complex and the overall survival was not satisfying. It was urgent to uncover novel molecules that play vital role in HNSCC for disease monitoring and drug development. METHODS Distinguished expression of FCGBP mRNA in HNSCC was analyzed by TCGA-HNSC and three GEO datasets, the relationship between FCGBP and clinical stage and survival was analyzed by GEPIA 2, the immune infiltration pattern analysis was conducted by TIMER 2.0, pathways affected by FCGBP was conducted by GSEA and GO/KEGG. In vitro experiments (including qRT-PCR, siRNA transfection, CCK8, transwell assay and flow cytometry) were conducted to confirm bioinformatic analysis. RESULTS FCGBP was down-regulated in tumor samples compared with normal tissues at both mRNA and protein levels, and positively correlated with survival in HNSCC. Genes co-expressed with FCGBP were mainly enriched in immune-related biological processes and pathways. GSEA indicated that FCGBP was associated with activated immune reaction and inhibiting well-known pro-tumor pathways. GSE41613 validated FCGBP as an independent prognostic marker for HNSCC and FCGBP was down-regulated in HNSCC cell lines by qRT-PCR. Migration and invasion of SCC9 and CAL27 were enhanced by FCGBP-targeting siRNAs, the ratio of cytotoxic T lymphocytes were down-regulated while the ratio of myeloid-derived suppressor cells were increased by FCGBP-targeting siRNAs. CONCLUSION FCGBP was a tumor suppressor gene and was an independent prognostic marker for better survival. The underlying mechanism may be that FCGBP inhibited tumor migration and invasion and activated immune response against tumor cells.
Collapse
Affiliation(s)
- Lijuan Zeng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Jun Zeng
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Department of General Dentistry and Oral Emergency, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Jianfeng He
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Yongqi Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Chengwei Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Zhiyan Lin
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Guangwei Chen
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Huilin Wu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China
| | - Libin Zhou
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China.
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, 195 Dongfengxi Road, Yuexiu District, Guangzhou, China.
| |
Collapse
|
4
|
Sheikh A, Ganguli D, Vickers TJ, Singer BB, Foulke-Abel J, Akhtar M, Khatoon N, Setu B, Basu S, Harro C, Maier N, Beatty WL, Chakraborty S, Bhuiyan TR, Qadri F, Donowitz M, Fleckenstein JM. Host-derived CEACAM-laden vesicles engage enterotoxigenic Escherichia coli for elimination and toxin neutralization. Proc Natl Acad Sci U S A 2024; 121:e2410679121. [PMID: 39264739 PMCID: PMC11420188 DOI: 10.1073/pnas.2410679121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/10/2024] [Indexed: 09/14/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of diarrheal illnesses annually ranging from mildly symptomatic cases to severe, life-threatening cholera-like diarrhea. Although ETEC are associated with long-term sequelae including malnutrition, the acute diarrheal illness is largely self-limited. Recent studies indicate that in addition to causing diarrhea, the ETEC heat-labile toxin (LT) modulates the expression of many genes in intestinal epithelia, including carcinoembryonic cell adhesion molecules (CEACAMs) which ETEC exploit as receptors, enabling toxin delivery. Here, however, we demonstrate that LT also enhances the expression of CEACAMs on extracellular vesicles (EV) shed by intestinal epithelia and that CEACAM-laden EV increase in abundance during human infections, mitigate pathogen-host interactions, scavenge free ETEC toxins, and accelerate ETEC clearance from the gastrointestinal tract. Collectively, these findings indicate that CEACAMs play a multifaceted role in ETEC pathogen-host interactions, transiently favoring the pathogen, but ultimately contributing to innate responses that extinguish these common infections.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Debayan Ganguli
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Tim J. Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Bernhard B. Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, 45147Essen, Germany
| | - Jennifer Foulke-Abel
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Marjahan Akhtar
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
- Enteric and Respiratory. Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka1212, Bangladesh
| | - Nazia Khatoon
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Bipul Setu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Clayton Harro
- Division of Global Disease Epidemiology and Control with the Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD21205
| | - Nicole Maier
- Center for Vaccine Innovation and Access, PATH, Seattle, WA98121
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Subhra Chakraborty
- Division of Global Disease Epidemiology and Control with the Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD21205
| | - Taufiqur R. Bhuiyan
- Enteric and Respiratory. Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka1212, Bangladesh
| | - Firdausi Qadri
- Enteric and Respiratory. Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka1212, Bangladesh
| | - Mark Donowitz
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - James M. Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
- Medicine Service, Infectious Disease Section, Veterans Affairs Health Care System, Saint Louis, MO63106
| |
Collapse
|
5
|
Kong C, Yang M, Yue N, Zhang Y, Tian C, Wei D, Shi R, Yao J, Wang L, Li D. Restore Intestinal Barrier Integrity: An Approach for Inflammatory Bowel Disease Therapy. J Inflamm Res 2024; 17:5389-5413. [PMID: 39161679 PMCID: PMC11330754 DOI: 10.2147/jir.s470520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
The intestinal barrier maintained by various types of columnar epithelial cells, plays a crucial role in regulating the interactions between the intestinal contents (such as the intestinal microbiota), the immune system, and other components. Dysfunction of the intestinal mucosa is a significant pathophysiological mechanism and clinical manifestation of inflammatory bowel disease (IBD). However, current therapies for IBD primarily focus on suppressing inflammation, and no disease-modifying treatments specifically target the epithelial barrier. Given the side effects associated with chronic immunotherapy, effective alternative therapies that promote mucosal healing are highly attractive. In this review, we examined the function of intestinal epithelial barrier function and the mechanisms of behind its disruption in IBD. We illustrated the complex process of intestinal mucosal healing and proposed therapeutic approaches to promote mucosal healing strategies in IBD. These included the application of stem cell transplantation and organ-like tissue engineering approaches to generate new intestinal tissue. Finally, we discussed potential strategies to restore the function of the intestinal barrier as a treatment for IBD.
Collapse
Affiliation(s)
- Chen Kong
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Meifeng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Ningning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Chengmei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Daoru Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Ruiyue Shi
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Jun Yao
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Lisheng Wang
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| | - Defeng Li
- The Second Clinical Medical College, Jinan University; Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
6
|
Sheikh A, Ganguli D, Vickers TJ, Singer B, Foulke-Abel J, Akhtar M, Khatoon N, Setu B, Basu S, Harro C, Maier N, Beatty WL, Chakraborty S, Bhuiyan TR, Qadri F, Donowitz M, Fleckenstein JM. Host-derived CEACAM-laden vesicles engage enterotoxigenic E. coli for elimination and toxin neutralization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604983. [PMID: 39091797 PMCID: PMC11291149 DOI: 10.1101/2024.07.24.604983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of diarrheal illnesses annually ranging from mildly symptomatic cases to severe, life-threatening cholera-like diarrhea. Although ETEC are associated with long-term sequelae including malnutrition, the acute diarrheal illness is largely self-limited. Recent studies indicate that in addition to causing diarrhea, the ETEC heat-labile toxin (LT) modulates the expression of many genes in intestinal epithelia, including carcinoembryonic cell adhesion molecules (CEACAMs) which ETEC exploit as receptors, enabling toxin delivery. Here however, we demonstrate that LT also enhances the expression of CEACAMs on extracellular vesicles (EV) shed by intestinal epithelia and that CEACAM-laden EV increase in abundance during human infections, mitigate pathogen-host interactions, scavenge free ETEC toxins, and accelerate ETEC clearance from the gastrointestinal tract. Collectively, these findings indicate that CEACAMs play a multifaceted role in ETEC pathogen-host interactions, transiently favoring the pathogen, but ultimately contributing to innate responses that extinguish these common infections.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Debayan Ganguli
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Tim J. Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Bernhard Singer
- Institute of Anatomy, Medical Faculty, University of Suisberg-Essen, 45147 Essen, Germany
| | - Jennifer Foulke-Abel
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marjahan Akhtar
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
- International Centre for Diarrhoeal Disease Research, Bangladesh; Dhaka, Bangladesh
| | - Nazia Khatoon
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Bipul Setu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Clayton Harro
- Department of International Health, Division of Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health
| | | | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Subhra Chakraborty
- Department of International Health, Division of Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health
| | - Tafiqur R. Bhuiyan
- International Centre for Diarrhoeal Disease Research, Bangladesh; Dhaka, Bangladesh
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh; Dhaka, Bangladesh
| | - Mark Donowitz
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James M. Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
- Medicine Service, Infectious Disease Section, Veterans Affairs Health Care System, Saint Louis, Missouri, USA
| |
Collapse
|
7
|
Yeshaya N, Gupta PK, Dym O, Morgenstern D, Major DT, Fass D. VWD domain stabilization by autocatalytic Asp-Pro cleavage. Protein Sci 2024; 33:e4929. [PMID: 38380729 PMCID: PMC10880436 DOI: 10.1002/pro.4929] [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: 09/25/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Domains known as von Willebrand factor type D (VWD) are found in extracellular and cell-surface proteins including von Willebrand factor, mucins, and various signaling molecules and receptors. Many VWD domains have a glycine-aspartate-proline-histidine (GDPH) amino-acid sequence motif, which is hydrolytically cleaved post-translationally between the aspartate (Asp) and proline (Pro). The Fc IgG binding protein (FCGBP), found in intestinal mucus secretions and other extracellular environments, contains 13 VWD domains, 11 of which have a GDPH cleavage site. In this study, we investigated the structural and biophysical consequences of Asp-Pro peptide cleavage in a representative FCGBP VWD domain. We found that endogenous Asp-Pro cleavage increases the resistance of the domain to exogenous proteolytic degradation. Tertiary structural interactions made by the newly generated chain termini, as revealed by a crystal structure of an FCGBP segment containing the VWD domain, may explain this observation. Notably, the Gly-Asp peptide bond, upstream of the cleavage site, assumed the cis configuration in the structure. In addition to these local features of the cleavage site, a global organizational difference was seen when comparing the FCGBP segment structure with the numerous other structures containing the same set of domains. Together, these data illuminate the outcome of GDPH cleavage and demonstrate the plasticity of proteins with VWD domains, which may contribute to their evolution for function in a dynamic extracellular environment.
Collapse
Affiliation(s)
- Noa Yeshaya
- Department of Chemical and Structural BiologyWeizmann Institute of ScienceRehovotIsrael
| | - Prashant Kumar Gupta
- Department of Chemistry and Institute for Nanotechnology & Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| | - Orly Dym
- Department of Life Sciences Core FacilitiesWeizmann Institute of ScienceRehovotIsrael
| | - David Morgenstern
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized MedicineWeizmann Institute of ScienceRehovotIsrael
| | - Dan Thomas Major
- Department of Chemistry and Institute for Nanotechnology & Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| | - Deborah Fass
- Department of Chemical and Structural BiologyWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
8
|
Hensel IV, Éliás S, Steinhauer M, Stoll B, Benfatto S, Merkt W, Krienke S, Lorenz HM, Haas J, Wildemann B, Resnik-Docampo M. SLE serum induces altered goblet cell differentiation and leakiness in human intestinal organoids. EMBO Mol Med 2024; 16:547-574. [PMID: 38316934 PMCID: PMC10940301 DOI: 10.1038/s44321-024-00023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024] Open
Abstract
Human intestinal epithelial cells are the interface between luminal content and basally residing immune cells. They form a tight monolayer that constantly secretes mucus creating a multilayered protective barrier. Alterations in this barrier can lead to increased permeability which is common in systemic lupus erythematosus (SLE) patients. However, it remains unexplored how the barrier is affected. Here, we present an in vitro model specifically designed to examine the effects of SLE on epithelial cells. We utilize human colon organoids that are stimulated with serum from SLE patients. Combining transcriptomic with functional analyses revealed that SLE serum induced an expression profile marked by a reduction of goblet cell markers and changed mucus composition. In addition, organoids exhibited imbalanced cellular composition along with enhanced permeability, altered mitochondrial function, and an interferon gene signature. Similarly, transcriptomic analysis of SLE colon biopsies revealed a downregulation of secretory markers. Our work uncovers a crucial connection between SLE and intestinal homeostasis that might be promoted in vivo through the blood, offering insights into the causal connection of barrier dysfunction and autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Wolfgang Merkt
- Division of Rheumatology, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Krienke
- Division of Rheumatology, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Haas
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
9
|
Guillén N. Pathogenicity and virulence of Entamoeba histolytica, the agent of amoebiasis. Virulence 2023; 14:2158656. [PMID: 36519347 DOI: 10.1080/21505594.2022.2158656] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
The amoeba parasite Entamoeba histolytica is the causative agent of human amebiasis, an enteropathic disease affecting millions of people worldwide. This ancient protozoan is an elementary example of how parasites evolve with humans, e.g. taking advantage of multiple mechanisms to evade immune responses, interacting with microbiota for nutritional and protective needs, utilizing host resources for growth, division, and encystation. These skills of E. histolytica perpetuate the species and incidence of infection. However, in 10% of infected cases, the parasite turns into a pathogen; the host-parasite equilibrium is then disorganized, and the simple lifecycle based on two cell forms, trophozoites and cysts, becomes unbalanced. Trophozoites acquire a virulent phenotype which, when non-controlled, leads to intestinal invasion with the onset of amoebiasis symptoms. Virulent E. histolytica must cross mucus, epithelium, connective tissue and possibly blood. This highly mobile parasite faces various stresses and a powerful host immune response, with oxidative stress being a challenge for its survival. New emerging research avenues and omics technologies target gene regulation to determine human or parasitic factors activated upon infection, their role in virulence activation, and in pathogenesis; this research bears in mind that E. histolytica is a resident of the complex intestinal ecosystem. The goal is to eradicate amoebiasis from the planet, but the parasitic life of E. histolytica is ancient and complex and will likely continue to evolve with humans. Advances in these topics are summarized here.
Collapse
Affiliation(s)
- Nancy Guillén
- Cell Biology and Infection Department, Institut Pasteur and Centre National de la Recherche Scientifique CNRS-ERM9195, Paris, France
| |
Collapse
|
10
|
Vornewald PM, Forman R, Yao R, Parmar N, Lindholm HT, Lee LSK, Martín-Alonso M, Else KJ, Oudhoff MJ. Mmp17-deficient mice exhibit heightened goblet cell effector expression in the colon and increased resistance to chronic Trichuris muris infection. Front Immunol 2023; 14:1243528. [PMID: 37869014 PMCID: PMC10587605 DOI: 10.3389/fimmu.2023.1243528] [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: 06/20/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Intestinal epithelial homeostasis is maintained by intrinsic and extrinsic signals. The extrinsic signals include those provided by mesenchymal cell populations that surround intestinal crypts and is further facilitated by the extracellular matrix (ECM), which is modulated by proteases such as matrix metalloproteinases (MMPs). Extrinsic signals ensure an appropriate balance between intestinal epithelial proliferation and differentiation. This study explores the role of MMP17, which is preferentially expressed by smooth muscle cells in the intestine, in intestinal homeostasis and during immunity to infection. Mice lacking MMP17 expressed high levels of goblet-cell associated genes and proteins, such as CLCA1 and RELM-β, which are normally associated with immune responses to infection. Nevertheless, Mmp17 KO mice did not have altered resistance during a bacterial Citrobacter rodentium infection. However, when challenged with a low dose of the helminth Trichuris muris, Mmp17 KO mice had increased resistance, without a clear role for an altered immune response during infection. Mechanistically, we did not find changes in traditional modulators of goblet cell effectors such as the NOTCH pathway or specific cytokines. We found MMP17 expression in smooth muscle cells as well as lamina propria cells such as macrophages. Together, our data suggest that MMP17 extrinsically alters goblet cell maturation which is sufficient to alter clearance in a helminth infection model.
Collapse
Affiliation(s)
- Pia M. Vornewald
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Ruth Forman
- Lydia Becker Institute of Immunology & Inflammation, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Rouan Yao
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Naveen Parmar
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard T. Lindholm
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Lilith S. K. Lee
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Mara Martín-Alonso
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
| | - Kathryn J. Else
- Lydia Becker Institute of Immunology & Inflammation, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Menno J. Oudhoff
- CEMIR – Center of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU – Norwegian University of Science and Technology, Trondheim, Norway
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
11
|
Wang CM, Fernez MT, Woolston BM, Carrier RL. Native gastrointestinal mucus: Critical features and techniques for studying interactions with drugs, drug carriers, and bacteria. Adv Drug Deliv Rev 2023; 200:114966. [PMID: 37329985 PMCID: PMC11184232 DOI: 10.1016/j.addr.2023.114966] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Gastrointestinal mucus plays essential roles in modulating interactions between intestinal lumen contents, including orally delivered drug carriers and the gut microbiome, and underlying epithelial and immune tissues and cells. This review is focused on the properties of and methods for studying native gastrointestinal mucus and its interactions with intestinal lumen contents, including drug delivery systems, drugs, and bacteria. The properties of gastrointestinal mucus important to consider in its analysis are first presented, followed by a discussion of different experimental setups used to study gastrointestinal mucus. Applications of native intestinal mucus are then described, including experimental methods used to study mucus as a barrier to drug delivery and interactions with intestinal lumen contents that impact barrier properties. Given the significance of the microbiota in health and disease, its impact on drug delivery and drug metabolism, and the use of probiotics and microbe-based delivery systems, analysis of interactions of bacteria with native intestinal mucus is then reviewed. Specifically, bacteria adhesion to, motility within, and degradation of mucus is discussed. Literature noted is focused largely on applications of native intestinal mucus models as opposed to isolated mucins or reconstituted mucin gels.
Collapse
Affiliation(s)
- Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Matthew T Fernez
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Benjamin M Woolston
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, USA; Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
12
|
Arnesen H, Markussen T, Birchenough G, Birkeland S, Nyström EEL, Hansson GC, Carlsen H, Boysen P. Microbial experience through housing in a farmyard-type environment alters intestinal barrier properties in mouse colons. Sci Rep 2023; 13:13701. [PMID: 37607995 PMCID: PMC10444815 DOI: 10.1038/s41598-023-40640-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
To close the gap between ultra-hygienic research mouse models and the much more environmentally exposed conditions of humans, we have established a system where laboratory mice are raised under a full set of environmental factors present in a naturalistic, farmyard-type habitat-a process we have called feralization. In previous studies we have shown that feralized (Fer) mice were protected against colorectal cancer when compared to conventionally reared laboratory mice (Lab). However, the protective mechanisms remain to be elucidated. Disruption of the protective intestinal barrier is an acknowledged player in colorectal carcinogenesis, and in the current study we assessed colonic mucosal barrier properties in healthy, feralized C57BL/6JRj male mice. While we found no effect of feralization on mucus layer properties, higher expression of genes encoding the mucus components Fcgbp and Clca1 still suggested mucus enforcement due to feralization. Genes encoding other proteins known to be involved in bacterial defense (Itln1, Ang1, Retnlb) and inflammatory mechanisms (Zbp1, Gsdmc2) were also higher expressed in feralized mice, further suggesting that the Fer mice have an altered intestinal mucosal barrier. These findings demonstrate that microbial experience conferred by housing in a farmyard-type environment alters the intestinal barrier properties in mice possibly leading to a more robust protection against disease. Future studies to unravel regulatory roles of feralization on intestinal barrier should aim to conduct proteomic analyses and in vivo performance of the feralized mice intestinal barrier.
Collapse
Affiliation(s)
- Henriette Arnesen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Turhan Markussen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - George Birchenough
- Mucin Biology Group, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Signe Birkeland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Elisabeth E L Nyström
- Mucin Biology Group, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C Hansson
- Mucin Biology Group, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Preben Boysen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway.
| |
Collapse
|
13
|
Gorman H, Moreau F, Dufour A, Chadee K. IgGFc-binding protein and MUC2 mucin produced by colonic goblet-like cells spatially interact non-covalently and regulate wound healing. Front Immunol 2023; 14:1211336. [PMID: 37359538 PMCID: PMC10285406 DOI: 10.3389/fimmu.2023.1211336] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
The colonic mucus bilayer is the first line of innate host defense that at the same time houses and nourishes the commensal microbiota. The major components of mucus secreted by goblet cells are MUC2 mucin and the mucus-associated protein, FCGBP (IgGFc-binding protein). In this study, we determine if FCGBP and MUC2 mucin were biosynthesized and interacted together to spatially enhance the structural integrity of secreted mucus and its role in epithelial barrier function. MUC2 and FCGBP were coordinately regulated temporally in goblet-like cells and in response to a mucus secretagogue but not in CRISPR-Cas9 gene-edited MUC2 KO cells. Whereas ~85% of MUC2 was colocalized with FCGBP in mucin granules, ~50% of FCGBP was diffusely distributed in the cytoplasm of goblet-like cells. STRING-db v11 analysis of the mucin granule proteome revealed no protein-protein interaction between MUC2 and FCGBP. However, FCGBP interacted with other mucus-associated proteins. FCGBP and MUC2 interacted via N-linked glycans and were non-covalently bound in secreted mucus with cleaved low molecular weight FCGBP fragments. In MUC2 KO, cytoplasmic FCGBP was significantly increased and diffusely distributed in wounded cells that healed by enhanced proliferation and migration within 2 days, whereas, in WT cells, MUC2 and FCGBP were highly polarized at the wound margin which impeded wound closure by 6 days. In DSS colitis, restitution and healed lesions in Muc2+/+ but not Muc2-/- littermates, were accompanied by a rapid increase in Fcgbp mRNA and delayed protein expression at 12- and 15-days post DSS, implicating a potential novel endogenous protective role for FCGBP in wound healing to maintain epithelial barrier function.
Collapse
Affiliation(s)
- Hayley Gorman
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - France Moreau
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Bian X, Si Z, Wang Q, Liu L, Shi Z, Tian C, Lee W, Zhang Y. IgG Fc-binding protein positively regulates the assembly of pore-forming protein complex βγ-CAT evolved to drive cell vesicular delivery and transport. J Biol Chem 2023; 299:104717. [PMID: 37068610 DOI: 10.1016/j.jbc.2023.104717] [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: 09/15/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023] Open
Abstract
Cell membranes form barriers for molecule exchange between the cytosol and the extracellular environments. βγ-CAT, a complex of pore-forming protein (PFP) BmALP1 (two βγ-crystallin domains with an aerolysin pore-forming domain) and the trefoil factor BmTFF3, has been identified in toad Bombina maxima. It plays pivotal roles, via inducing channel formation in various intra- or extra- cellular vesicles, as well as in nutrient acquisition, maintaining water balance, and antigen presentation. Thus, such a protein machine should be tightly regulated. Indeed, BmALP3 (a paralog of BmALP1) oxidizes BmALP1 to form a water-soluble polymer, leading to dissociation of the βγ-CAT complex and loss of biological activity. Here, we found that the B. maxima IgG Fc-binding protein (FCGBP), a well-conserved vertebrate mucin-like protein with unknown functions, acted as a positive regulator for βγ-CAT complex assembly. The interactions among FCGBP, BmALP1, and BmTFF3 were revealed by co-immunoprecipitation assays. Interestingly, FCGBP reversed the inhibitory effect of BmALP3 on the βγ-CAT complex. Furthermore, FCGBP reduced BmALP1 polymers and facilitated the assembly of βγ-CAT with the biological pore-forming activity in the presence of BmTFF3. Our findings define the role of FCGBP in mediating the assembly of a PFP machine evolved to drive cell vesicular delivery and transport.
Collapse
Affiliation(s)
- Xianling Bian
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Ziru Si
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Qiquan Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Human Aging Research Institute (HARI) and School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Lingzhen Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Zhihong Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Changlin Tian
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Wenhui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences/Engineering Laboratory of Peptides of the Chinese Academy of Sciences, Institute of Zoology, the Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
15
|
McDonald CA, Becker CG, Lambertini C, Toledo LF, Haddad CFB, Zamudio KR. Host immune responses to enzootic and invasive pathogen lineages vary in magnitude, timing, and efficacy. Mol Ecol 2023; 32:2252-2270. [PMID: 36799008 DOI: 10.1111/mec.16890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Infectious diseases of wildlife continue to pose a threat to biodiversity worldwide, yet pathogens are far from uniform in virulence or host disease outcome. Within the same pathogen species, virulence can vary considerably depending on strain or lineage, in turn eliciting variable host responses. One pathogen that has caused extensive biodiversity loss is the amphibian-killing fungus, Batrachochytrium dendrobatidis (Bd), which is comprised of a globally widespread hypervirulent lineage (Bd-GPL), and multiple geographically restricted, enzootic lineages. Whereas host immunogenomic responses to Bd-GPL have been characterized in a number of amphibian species, immunogenomic responses to geographically restricted, enzootic Bd lineages are less clear. To examine lineage-specific host immune responses to Bd, we exposed a species of pumpkin toadlet, Brachycephalus pitanga, which is endemic to Brazil's Southern Atlantic Forest, to either the Bd-GPL or the enzootic Bd-Asia-2/Brazil (hereafter Bd-Brazil) lineage. Using temporal samples from early, mid, and late infection stages, we quantified functional immunogenomic responses over the course of infection using differential gene expression tests and coexpression network analyses. Host immune responses varied significantly with Bd lineage. Relative to controls, toadlet responses to Bd-Brazil were weak at early infection (25 genes significantly differentially expressed), peaked by mid-stage infection (414 genes), and were nearly fully resolved by late-stage infection (nine genes). In contrast, responses to Bd-GPL were magnified and delayed; toadlets significantly differentially expressed 111 genes early, 87 genes at mid-stage infection, and 726 genes by late-stage infection relative to controls. Given that infection intensity did not vary between mid- and late-stage disease in either Bd-Brazil or Bd-GPL treatments, this suggests that pumpkin toadlets may be at least partially tolerant to the enzootic Bd-Brazil lineage. In contrast, late-stage immune activation against Bd-GPL was consistent with immune dysregulation previously observed in other species. Our results demonstrate that both the timing of immune response and the particular immune pathways activated are specific to Bd lineage. Within regions where multiple Bd lineages co-occur, and given continued global Bd movement, these differential host responses may influence not only individual disease outcome, but transmission dynamics at the population and community levels.
Collapse
Affiliation(s)
- Coby A McDonald
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - C Guilherme Becker
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Carolina Lambertini
- Laboratório de História Natural de Anfíbios Brasileiros (LaHNAB), Departamento de Biologia Animal, Instituto de Biologia Unicamp, Campinas, São Paulo, Brazil
| | - L Felipe Toledo
- Laboratório de História Natural de Anfíbios Brasileiros (LaHNAB), Departamento de Biologia Animal, Instituto de Biologia Unicamp, Campinas, São Paulo, Brazil
| | - Célio F B Haddad
- Departamento de Biodiversidade e Centro de Aquicultura (CAUNESP), Instituto de Biociências, Universidade Estadual Paulista, Rio Claro, Brazil
| | - Kelly R Zamudio
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA.,Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
16
|
Weste J, Houben T, Harder S, Schlüter H, Lücke E, Schreiber J, Hoffmann W. Different Molecular Forms of TFF3 in the Human Respiratory Tract: Heterodimerization with IgG Fc Binding Protein (FCGBP) and Proteolytic Cleavage in Bronchial Secretions. Int J Mol Sci 2022; 23:ijms232315359. [PMID: 36499686 PMCID: PMC9737082 DOI: 10.3390/ijms232315359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The polypeptide TFF3 belongs to the trefoil factor family (TFF) of lectins. TFF3 is typically secreted from mucous epithelia together with mucins. Both intestinal and salivary TFF3 mainly exist as disulfide-linked heterodimers with IgG Fc binding protein (FCGBP). Here, we investigated bronchial tissue specimens, bronchial secretions, and bronchoalveolar lavage (BAL) fluid from patients with a chronic obstructive pulmonary disease (COPD) background by fast protein liquid chromatography and proteomics. For the first time, we identified different molecular forms of TFF3 in the lung. The high-molecular mass form represents TFF3-FCGBP oligomers, whereas the low-molecular mass forms are homodimeric and monomeric TFF3 with possibly anti-apoptotic activities. In addition, disulfide-linked TFF3 heterodimers with an Mr of about 60k and 30k were detected in both bronchial secretions and BAL fluid. In these liquids, TFF3 is partly N-terminally truncated probably by neutrophil elastase cleavage. TFF3-FCGBP is likely involved in the mucosal innate immune defense against microbial infections. We discuss a hypothetical model how TFF3 might control FCGBP oligomerization. Furthermore, we did not find indications for interactions of TFF3-FCGBP with DMBT1gp340 or the mucin MUC5AC, glycoproteins involved in mucosal innate immunity. Surprisingly, bronchial MUC5AC appeared to be degraded when compared with gastric MUC5AC.
Collapse
Affiliation(s)
- Jens Weste
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Till Houben
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sönke Harder
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eva Lücke
- Department of Pneumology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Jens Schreiber
- Department of Pneumology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
17
|
Gustafsson JK, Johansson MEV. The role of goblet cells and mucus in intestinal homeostasis. Nat Rev Gastroenterol Hepatol 2022; 19:785-803. [PMID: 36097076 DOI: 10.1038/s41575-022-00675-x] [Citation(s) in RCA: 257] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/08/2022]
Abstract
The intestinal tract faces numerous challenges that require several layers of defence. The tight epithelium forms a physical barrier that is further protected by a mucus layer, which provides various site-specific protective functions. Mucus is produced by goblet cells, and as a result of single-cell RNA sequencing identifying novel goblet cell subpopulations, our understanding of their various contributions to intestinal homeostasis has improved. Goblet cells not only produce mucus but also are intimately linked to the immune system. Mucus and goblet cell development is tightly regulated during early life and synchronized with microbial colonization. Dysregulation of the developing mucus systems and goblet cells has been associated with infectious and inflammatory conditions and predisposition to chronic disease later in life. Dysfunctional mucus and altered goblet cell profiles are associated with inflammatory conditions in which some mucus system impairments precede inflammation, indicating a role in pathogenesis. In this Review, we present an overview of the current understanding of the role of goblet cells and the mucus layer in maintaining intestinal health during steady-state and how alterations to these systems contribute to inflammatory and infectious disease.
Collapse
Affiliation(s)
- Jenny K Gustafsson
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemisty and Cell biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
18
|
Liu Q, Niu X, Li Y, Zhang JR, Zhu SJ, Yang QY, Zhang W, Gong L. Role of the mucin-like glycoprotein FCGBP in mucosal immunity and cancer. Front Immunol 2022; 13:863317. [PMID: 35936008 PMCID: PMC9354016 DOI: 10.3389/fimmu.2022.863317] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/27/2022] [Indexed: 12/26/2022] Open
Abstract
IgGFc-binding protein (FCGBP) is a mucin first detected in the intestinal epithelium. It plays an important role in innate mucosal epithelial defense, tumor metastasis, and tumor immunity. FCGBP forms disulfide-linked heterodimers with mucin-2 and members of the trefoil factor family. These formed complexes inhibit bacterial attachment to mucosal surfaces, affect the motility of pathogens, and support their clearance. Altered FCGBP expression levels may be important in the pathologic processes of Crohn’s disease and ulcerative colitis. FCGBP is also involved in regulating the infiltration of immune cells into tumor microenvironments. Thus, the molecule is a valuable marker of tumor prognosis. This review summarizes the functional relevance and role of FCGBP in immune responses and disease development, and highlights the potential role in diagnosis and predicting tumor prognosis.
Collapse
Affiliation(s)
- Qiao Liu
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Xia Niu
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Yang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jia-rui Zhang
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Shao-jun Zhu
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Qi-yuan Yang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Wei Zhang
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
- *Correspondence: Li Gong, ; Wei Zhang,
| | - Li Gong
- Department of Pathology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
- *Correspondence: Li Gong, ; Wei Zhang,
| |
Collapse
|
19
|
Fang J, Zhang Z, Cheng Y, Yang H, Zhang H, Xue Z, Lu S, Dong Y, Song C, Zhang X, Zhou Y. EPA and DHA differentially coordinate the crosstalk between host and gut microbiota and block DSS-induced colitis in mice by a reinforced colonic mucus barrier. Food Funct 2022; 13:4399-4420. [PMID: 35297435 DOI: 10.1039/d1fo03815j] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background: Ulcerative colitis (UC) is a chronic inflammatory disorder of the colon with a continuously remitting and relapsing course. Its etiology is closely related to abnormal interactions between host and gut microbiota. The mucus barrier lining the gastrointestinal tract is necessary to coordinate host and gut microbiota interaction by nourishing and modulating the microbiota. Differential effects of the anti-inflammatory fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on UC progression in mice were firstly addressed by our previous work; here, the mechanism for their respective effects were further uncovered from host-microbiome crosstalk based on mucus barrier modulation to pave the way for UC therapy. Methods: Assessment of the disease activity index and histopathology score was conducted in mice with dextran sodium sulfate (DSS)-induced colitis pre-treated with different doses of EPA and DHA. Mucin generation, glycosylation and secretion were evaluated by a combination of electron microscopy, specific mucous staining, and qPCR. Western blotting was used to analyze the underlying molecular events. Fecal short chain fatty acids were detected using gas chromatography, and the gut microbial composition was analyzed using 16S rRNA sequencing. Results: Compared with DHA, the more potent inhibitory effect of high dose EPA on DSS-induced colitis was reconfirmed, which was underlain by a reinforced mucus layer as indicated by increased mucin granule release, mucus layer stratification and markedly upregulated expression of the key modulators involved in goblet cell differentiation. In turn a remarkably enhanced mucus barrier in the EPA group functioned to modulate the gut microbiome, as demonstrated by the enriched abundance of the phylum Bacteroidetes and mucin-degrading bacterium Akkermansia muciniphila producing acetic and propionic acids. Conclusions: EPA and DHA differentially coordinate the interaction between the host and the gut microbiota and relieve mucus barrier disruption in DSS-induced colitis. EPA may develop into a promising adjunctive therapy for UC.
Collapse
Affiliation(s)
- Jian Fang
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,College of Medicine, Shaoxing University, 508 Huancheng Road, Shaoxing, Zhejiang Province, 312000, People's Republic of China
| | - ZhuangWei Zhang
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Yinyin Cheng
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Haitao Yang
- Mingzhou Hospital of Zhejiang University Department of Pathology, Mingzhou Hospital of Zhejiang University, Ningbo, 315040 Zhejiang, People's Republic of China
| | - Hui Zhang
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Zhe Xue
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Songtao Lu
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Yichen Dong
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Chunyan Song
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Xiaohong Zhang
- Department of Preventive Medicine, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo, Zhejiang, 315211, People's Republic of China.,Department of Gastroenterology and hepatology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yuping Zhou
- Department of Gastroenterology and hepatology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Ningbo, Zhejiang, 315020, People's Republic of China. .,Institute of Digestive Disease of Ningbo University, Ningbo, 315020, People's Republic of China
| |
Collapse
|
20
|
Abstract
The immune system in the large intestine is separated from commensal microbes and comparatively rare enteric pathogens by a monolayer of diverse epithelial cells overlaid with a compact and adherent inner mucus layer and a looser outer mucus layer. Microorganisms, collectively referred to as the mucus-associated (MA) microbiota, physically inhabit this mucus barrier, resulting in a dynamic and incessant dialog to maintain both spatial segregation and immune tolerance. Recent major findings reveal novel features of the crosstalk between the immune system and mucus-associated bacteria in health and disease, as well as disease-related peripheral immune signatures indicative of host responses to these organisms. In this brief review, we integrate these novel observations into our overall understanding of host-microbiota mutualism at the colonic mucosal border and speculate on the significance of this emerging knowledge for our understanding of the prevention, development, and progression of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Medicine, University of Alabama at Birmingham, Birmingham, 35294, USA
| | - Craig L. Maynard
- Department of Medicine, University of Alabama at Birmingham, Birmingham, 35294, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, 35294, USA
| |
Collapse
|
21
|
The Impact of MicroRNAs during Inflammatory Bowel Disease: Effects on the Mucus Layer and Intercellular Junctions for Gut Permeability. Cells 2021; 10:cells10123358. [PMID: 34943865 PMCID: PMC8699384 DOI: 10.3390/cells10123358] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/15/2022] Open
Abstract
Research on inflammatory bowel disease (IBD) has produced mounting evidence for the modulation of microRNAs (miRNAs) during pathogenesis. MiRNAs are small, non-coding RNAs that interfere with the translation of mRNAs. Their high stability in free circulation at various regions of the body allows researchers to utilise miRNAs as biomarkers and as a focus for potential treatments of IBD. Yet, their distinct regulatory roles at the gut epithelial barrier remain elusive due to the fact that there are several external and cellular factors contributing to gut permeability. This review focuses on how miRNAs may compromise two components of the gut epithelium that together form the initial physical barrier: the mucus layer and the intercellular epithelial junctions. Here, we summarise the impact of miRNAs on goblet cell secretion and mucin structure, along with the proper function of various junctional proteins involved in paracellular transport, cell adhesion and communication. Knowledge of how this elaborate network of cells at the gut epithelial barrier becomes compromised as a result of dysregulated miRNA expression, thereby contributing to the development of IBD, will support the generation of miRNA-associated biomarker panels and therapeutic strategies that detect and ameliorate gut permeability.
Collapse
|
22
|
Parrish A, Boudaud M, Kuehn A, Ollert M, Desai MS. Intestinal mucus barrier: a missing piece of the puzzle in food allergy. Trends Mol Med 2021; 28:36-50. [PMID: 34810087 DOI: 10.1016/j.molmed.2021.10.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022]
Abstract
The prevalence of food allergies has reached epidemic levels but the cause remains largely unknown. We discuss the clinical relevance of the gut mucosal barrier as a site for allergic sensitization to food. In this context, we focus on an important but overlooked part of the mucosal barrier in pathogenesis, the glycoprotein-rich mucus layer, and call attention to both beneficial and detrimental aspects of mucus-gut microbiome interactions. Studying the intricate links between the mucus barrier, the associated bacteria, and the mucosal immune system may advance our understanding of the mechanisms and inform prevention and treatment strategies in food allergy.
Collapse
Affiliation(s)
- Amy Parrish
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Annette Kuehn
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark.
| |
Collapse
|