1
|
Liu C, Shen Y, Yang M, Chi K, Guo N. Hazard of Staphylococcal Enterotoxins in Food and Promising Strategies for Natural Products against Virulence. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:2450-2465. [PMID: 35170308 DOI: 10.1021/acs.jafc.1c06773] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Staphylococcal enterotoxins (SEs) secreted by Staphylococcus aureus frequently contaminate food and cause serious foodborne diseases but are ignored during food processing and even cold-chain storage. Notably, SEs are stable and resistant to harsh sterilization environments, which can induce more serious hazards to public health than the bacterium itself. Therefore, it is necessary to develop promising strategies to control SE contamination in food and improve food safety. Natural products not only have various pharmaceutical properties, such as antimicrobial and antitoxin activities, but they are also eco-friendly, safe, nutritive, and barely drug-resistant. Here, the hazards of SEs and the promising natural compounds with different inhibitory mechanisms are summarized and classified. The key points of future research and applications for natural products against bacterial toxin contamination in food are also prospected. Overall, this review may provide enlightening insights for screening effective natural compounds to prevent foodborne diseases caused by bacterial toxins.
Collapse
Affiliation(s)
- Chunmei Liu
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun, Jilin 130062, People's Republic of China
| | - Yong Shen
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun, Jilin 130062, People's Republic of China
| | - Meng Yang
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun, Jilin 130062, People's Republic of China
| | - Kunmei Chi
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun, Jilin 130062, People's Republic of China
| | - Na Guo
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Road, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
2
|
Markandey M, Bajaj A, Ilott NE, Kedia S, Travis S, Powrie F, Ahuja V. Gut microbiota: sculptors of the intestinal stem cell niche in health and inflammatory bowel disease. Gut Microbes 2022; 13:1990827. [PMID: 34747326 PMCID: PMC8583176 DOI: 10.1080/19490976.2021.1990827] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Intestinal epithelium represents a dynamic and diverse cellular system that continuously interacts with gut commensals and external cues. Intestinal stem cells, which lie at the heart of epithelial renewal and turnover, proliferate to maintain a steady stem cell population and differentiate to form functional epithelial cell types. This rather sophisticated assembly-line is maintained by an elaborate micro-environment, sculpted by a myriad of host and gut microbiota-derived signals, forming an intestinal stem cell niche. This complex, yet crucial signaling niche undergoes dynamic changes during homeostasis and chronic intestinal inflammation. Inflammatory bowel disease refers to a chronic inflammatory response toward pathogenic or commensal microbiota, in a genetically susceptible host. Compositional and functional alterations in gut microbiota are pathognomonic of IBD.The present review highlights the modulatory role of gut microbiota on the intestinal stem cell niche during homeostasis and inflammatory bowel disease. We discuss the mechanisms of direct action of gut commensals (through microbiota-derived or microbiota-influenced metabolites) on ISCs, followed by their effects via other epithelial and immune cell types.
Collapse
Affiliation(s)
- Manasvini Markandey
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Aditya Bajaj
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | | | - Saurabh Kedia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Simon Travis
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Vineet Ahuja
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India,CONTACT Vineet Ahuja Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India, 110029
| |
Collapse
|
3
|
Almutawif Y, Hartmann B, Lloyd M, Lai CT, Rea A, Geddes D. Staphylococcus aureus Enterotoxin Production in Raw, Holder-Pasteurized, and Ultraviolet-C-Treated Donated Human Milk. Breastfeed Med 2019; 14:262-270. [PMID: 30817174 DOI: 10.1089/bfm.2018.0217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Some strains of Staphylococcus aureus can produce heat-stable enterotoxins that have been associated with gastritis and potentially necrotizing enterocolitis in preterm infants. Objectives/Hypothesis: To assess the impact of different storage temperatures on S. aureus growth and enterotoxin production in raw, Holder-pasteurized (HP) and ultraviolet-C (UV-C)-treated donated human milk (DHM). Materials and Methods: The milk samples from individual donors were pooled and divided into four equal portions. One portion was HP, the second was UV-C treated, the third was not treated, and the fourth was UV-C treated after being spiked with S. aureus. All samples were incubated at 37°C (18 hours) and 4°C (14 days). Bacterial colony count, enterotoxin A and B, and immune proteins were quantified. Results: At 37°C, the colony count increased in HP DHM and decreased in raw and UV-C-treated DHM. At 4°C, colony counts in HP DHM reduced and were not detected in raw and UV-C-treated DHM from day 8 of incubation. No bacteria were detected in samples that were inoculated before UV-C treatment. Enterotoxin A was only detected in HP-DHM at 37°C from the 9th hour onward. Enterotoxin B was detected in one sample at the 15th hour. Immune protein concentrations were similar in raw and UV-C DHM, and were reduced in the HP DHM. Conclusion: UV-C-treated milk reduces S. aureus growth with similar kinetics to raw milk making it a promising emerging technique to eliminate bacteria while retaining essential immune proteins in DHM.
Collapse
Affiliation(s)
- Yahya Almutawif
- 1 Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia.,2 School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia.,3 Marshall Center for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia
| | - Benjamin Hartmann
- 4 Perron Rotary Express Milk Bank, King Edward Memorial Hospital, Subiaco, Australia.,5 Center for Neonatal Research and Education, The University of Western Australia, Crawley, Australia.,6 School of Paediatrics, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia
| | - Megan Lloyd
- 2 School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia.,3 Marshall Center for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia.,6 School of Paediatrics, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia.,7 School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Ching Tat Lai
- 8 School of Molecular Sciences, Faculty of Science, The University of Western Australia, Crawley, Australia
| | - Alethea Rea
- 9 Center for Applied Statistics, The University of Western Australia, Crawley, Australia
| | - Donna Geddes
- 8 School of Molecular Sciences, Faculty of Science, The University of Western Australia, Crawley, Australia
| |
Collapse
|
4
|
Almutawif Y, Hartmann B, Lloyd M, Lai CT, Rea A, Geddes D. Staphylococcus aureus Enterotoxin Production in Raw and Pasteurized Milk: The Effect of Selected Different Storage Durations and Temperatures. Breastfeed Med 2019; 14:256-261. [PMID: 30844297 DOI: 10.1089/bfm.2018.0227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction:S. aureus is one of the most prevalent potential pathogenic bacteria found in DHM. Some strains produce heat stable enterotoxins that are able to survive pasteurization. These enterotoxins have been associated with gastritis and potentially necrotizing enterocolitis in preterm infants. The aim of this study is to assess the impact of different storage temperatures on Staphylococcus aureus growth and enterotoxin A and B production in raw and Holder pasteurized donor human milk (DHM). Materials and Methods: Raw and pasteurized DHM were inoculated with enterotoxin A and B producing S. aureus. Samples were incubated at 4°C (10 days), 21°C, and 37°C (18 hours). Bacterial growth and enterotoxin A and B were quantified. Results:S. aureus count increased in pasteurized DHM. Bacterial count decreased in the raw milk when incubated at 21°C and 4°C and slightly increased when incubated at 37°C. Enterotoxins A and B were only detected in pasteurized DHM at 37°C from 9 hours onward. Conclusion: This study showed that raw milk is capable of suppressing S. aureus growth compared to pasteurized DHM. It also provides a measure of assurance of the safety of raw and pasteurized DHM when refrigerated or left at room temperature for a few hours.
Collapse
Affiliation(s)
- Yahya Almutawif
- 1 Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia.,2 Faculty of Health and Medical Sciences, School of Biomedical Sciences, The University of Western Australia, Crawley, Australia.,3 Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia
| | - Benjamin Hartmann
- 4 Perron Rotary Express Milk Bank, King Edward Memorial Hospital, Subiaco, Australia.,5 Centre for Neonatal Research and Education, The University of Western Australia, Crawley, Australia.,6 School of Paediatrics, Faculty of Health Sciences, The University of Western Australia, Crawley, Australia
| | - Megan Lloyd
- 2 Faculty of Health and Medical Sciences, School of Biomedical Sciences, The University of Western Australia, Crawley, Australia.,3 Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia.,6 School of Paediatrics, Faculty of Health Sciences, The University of Western Australia, Crawley, Australia.,7 School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Ching Tat Lai
- 8 Faculty of Science, School of Molecular Sciences, The University of Western Australia (M310), Crawley, Australia
| | - Alethea Rea
- 9 Centre for Applied Statistics, The University of Western Australia, Crawley, Australia
| | - Donna Geddes
- 8 Faculty of Science, School of Molecular Sciences, The University of Western Australia (M310), Crawley, Australia
| |
Collapse
|
5
|
Beswick EJ, Grim C, Singh A, Aguirre JE, Tafoya M, Qiu S, Rogler G, McKee R, Samedi V, Ma TY, Reyes VE, Powell DW, Pinchuk IV. Expression of Programmed Death-Ligand 1 by Human Colonic CD90 + Stromal Cells Differs Between Ulcerative Colitis and Crohn's Disease and Determines Their Capacity to Suppress Th1 Cells. Front Immunol 2018; 9:1125. [PMID: 29910803 PMCID: PMC5992387 DOI: 10.3389/fimmu.2018.01125] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/03/2018] [Indexed: 12/14/2022] Open
Abstract
Background and Aims The role of programmed cell death protein 1 (PD-1) and its ligands in the dysregulation of T helper immune responses observed in the inflammatory bowel disease (IBD) is unclear. Recently, a novel concept emerged that CD90+ colonic (myo)fibroblasts (CMFs), also known as stromal cells, act as immunosuppressors, and are among the key regulators of acute and chronic inflammation. The objective of this study was to determine if the level of the PD-1 ligands is changed in the IBD inflamed colonic mucosa and to test the hypothesis that changes in IBD-CMF-mediated PD-1 ligand-linked immunosuppression is a mechanism promoting the dysregulation of Th1 cell responses. Methods Tissues and cells derived from Crohn's disease (CD), ulcerative colitis (UC), and healthy individuals (N) were studied in situ, ex vivo, and in culture. Results A significant increase in programmed death-ligand 1 (PD-L1) was observed in the inflamed UC colonic mucosa when compared to the non-inflamed matched tissue samples, CD, and healthy controls. UC-CMFs were among the major populations in the colonic mucosa contributing to the enhanced PD-L1 expression. In contrast, PD-L1 expression was decreased in CD-CMFs. When compared to CD-CMFs and N-CMFs, UC-CMFs demonstrated stronger suppression of IL-2, Th1 transcriptional factor Tbet, and IFN-γ expression by CD3/CD28-activated CD4+ T cells, and this process was PD-L1 dependent. Similar observations were made when differentiated Th1 cells were cocultured with UC-CMFs. In contrast, CD-CMFs showed reduced capacity to suppress Th1 cell activity and addition of recombinant PD-L1 Fc to CD-CMF:T cell cocultures partially restored the suppression of the Th1 type responses. Conclusion We present evidence showing that increased PD-L1 expression suppresses Th1 cell activity in UC. In contrast, loss of PD-L1 expression observed in CD contributes to the persistence of the Th1 inflammatory milieu in CD. Our data suggest that dysregulation of the Th1 responses in the inflamed colonic mucosa of IBD patients is promoted by the alterations in PD-L1 expression in the mucosal mesenchymal stromal cell compartment.
Collapse
Affiliation(s)
- Ellen J Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, United States
| | - Carl Grim
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Abinav Singh
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Jose E Aguirre
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Marissa Tafoya
- Department of Pathology, University of New Mexico, Albuquerque, NM, United States
| | - Suimin Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital of Zürich, Zürich, Switzerland
| | - Rohini McKee
- Department of Surgery, University of New Mexico, Albuquerque, NM, United States
| | - Von Samedi
- Department of Pathology, University of New Mexico, Albuquerque, NM, United States
| | - Thomas Y Ma
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM, United States
| | - Victor E Reyes
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States
| | - Don W Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Irina V Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
6
|
Pinchuk IV, Powell DW. Immunosuppression by Intestinal Stromal Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:115-129. [DOI: 10.1007/978-3-319-78127-3_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
7
|
Benkerroum N. Staphylococcal enterotoxins and enterotoxin-like toxins with special reference to dairy products: An overview. Crit Rev Food Sci Nutr 2017; 58:1943-1970. [DOI: 10.1080/10408398.2017.1289149] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Noreddine Benkerroum
- Department of Food Science and Agricultural Chemistry, Macdonald-Stewart Building, McGill University, Macdonald Campus, Sainte-Anne-de-Bellevue, Canada
| |
Collapse
|
8
|
Watts TL, Cui R, Szaniszlo P, Resto VA, Powell DW, Pinchuk IV. PDGF-AA mediates mesenchymal stromal cell chemotaxis to the head and neck squamous cell carcinoma tumor microenvironment. J Transl Med 2016; 14:337. [PMID: 27931212 PMCID: PMC5146849 DOI: 10.1186/s12967-016-1091-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/22/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The robust desmoplasia associated with head and neck squamous cell carcinoma (HNSCC) suggests that the tumor microenvironment may be an important component in the pathophysiology of this cancer. Moreover, the high recurrence rate and poor clinical response to chemotherapy and radiation treatment further underscores that the non-cancerous cells of the microenvironment, such as mesenchymal stromal cells (MSCs), cancer associated fibroblasts (CAFs), and pericytes, may be important in the pathophysiology of HNSCC. METHODS Confocal microscopy and immunohistomchemistry approaches were used to identify MSCs tumor microenvironment from patients with oral cavity and oral pharyngeal squamous cell carcinoma (SCC). In vitro Boyden chamber assays and multiplex magnetic bead assays were used to measure MSC chemotaxis and to identify the chemokines secreted by JHU-011, -012, -019, three cells lines derived from patients with oral pharyngeal SCC. RESULTS We show here that MSCs reside in the tumor microenvironment of patients with oral cavity and oral pharyngeal SCC and are recruited via paracrine mediated tumor cell secretion of (platelet derived growth factor) PDGF-AA. The MSC markers CD90+, CD105+, and gremlin-1+ were found to co-localize on cells within the tumor microenvironment in oral cavity SCC specimens distinct from α-smooth muscle actin staining CAFs. The conditioned media from JHU-011, -012, and -019 caused a significant increase in MSC migration (>60%) and invasion (>50%; p < 0.0001) compared to oral keratinocyte (OKT) controls. Tumor cell induced MSC chemotaxis appears to be mediated through paracrine secretion of PDGF-AA as inhibition of the PDGF-AA receptor, PDGFR-α but not PDGFR-β, resulted in near arrest of MSC chemotaxis (p < 0.0001). CONCLUSIONS Tumor microenvironment expression of PDGFR-α has been shown to correlate with a worse prognosis in patients with prostate, breast, ovarian, non-small cell lung cancer and osteosarcoma. This is the first evidence that a similar signaling paradigm may be present in HNSCC. PDGFR-α inhibitors have not been studied as adjunctive treatment options in the management of HNSCC and may prove to be an important driver of the malignant phenotype in this setting.
Collapse
Affiliation(s)
- Tammara L Watts
- Department of Otolaryngology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0521, USA.
| | - Ruwen Cui
- Department of Otolaryngology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0521, USA
| | - Peter Szaniszlo
- Department of Otolaryngology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0521, USA
| | - Vicente A Resto
- Department of Otolaryngology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555-0521, USA
| | - Don W Powell
- Internal Medicine, Division of Gastroenterology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Irina V Pinchuk
- Internal Medicine, Division of Gastroenterology, University of Texas Medical Branch, Galveston, TX, 77555, USA.,Microbiology and Immunology Department, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
9
|
Johnson P, Beswick EJ, Chao C, Powell DW, Hellmich MR, Pinchuk IV. Isolation of CD 90+ Fibroblast/Myofibroblasts from Human Frozen Gastrointestinal Specimens. J Vis Exp 2016:e53691. [PMID: 26863470 DOI: 10.3791/53691] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Fibroblasts/myofibroblasts (MFs) have been gaining increasing attention for their role in pathogenesis and their contributions to both wound healing and promotion of the tumor microenvironment. While there are currently many techniques for the isolation of MFs from gastrointestinal (GI) tissues, this protocol introduces a novel element of isolation of these stromal cells from frozen tissue. Freezing GI tissue specimens not only allows the researcher to acquire samples from worldwide collaborators, biobanks, and commercial vendors, it also permits the delayed processing of fresh samples. The described protocol will consistently yield characteristic spindle-shaped cells with the MF phenotype that express the markers CD90, α-SMA and vimentin. As these cells are derived from patient samples, the use of primary cells also confers the benefit of closely mimicking MFs from disease states-namely cancer and inflammatory bowel diseases. This technique has been validated in gastric, small bowel, and colonic MF primary culture generation. Primary MF cultures can be used in a vast array of experiments over a number of passage and their purity assessed by both immunocytochemistry and flow cytometry analysis.
Collapse
Affiliation(s)
| | - Ellen J Beswick
- Molecular Genetics and Microbiology, University of New Mexico
| | - Celia Chao
- Surgery, University of Texas Medical Branch
| | - Don W Powell
- Internal Medicine, University of Texas Medical Branch
| | | | | |
Collapse
|
10
|
Huynh PT, Beswick EJ, Coronado YA, Johnson P, O'Connell MR, Watts T, Singh P, Qiu S, Morris K, Powell DW, Pinchuk IV. CD90(+) stromal cells are the major source of IL-6, which supports cancer stem-like cells and inflammation in colorectal cancer. Int J Cancer 2015; 138:1971-81. [PMID: 26595254 DOI: 10.1002/ijc.29939] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 10/01/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022]
Abstract
IL-6 is a pleiotropic cytokine increased in CRC and known to directly promote tumor growth. Colonic myofibroblasts/fibroblasts (CMFs or stromal cells) are CD90(+) innate immune cells representing up to 30% of normal colonic mucosal lamina propria cells. They are expanded in CRC tumor stroma, where they also known as a cancer associated fibroblasts (CAFs). Cells of mesenchymal origin, such as normal myofibroblasts/fibroblasts, are known to secrete IL-6; however, their contribution to the increase in IL-6 in CRC and to tumor-promoting inflammation is not well defined. Using in situ, ex vivo and coculture analyses we have demonstrated that the number of IL-6 producing CMFs is increased in CRC (C-CMFs) and they represent the major source of IL-6 in T2-T3 CRC tumors. Activity/expression of stem cell markers-aldehyde dehydrogenase and LGR5- was significantly up-regulated in colon cancer cells (SW480, Caco-2 or HT29) cultured in the presence of conditioned medium from tumor isolated C-CMFs in an IL-6 dependent manner. C-CMF and its derived condition medium, but not normal CMF isolated from syngeneic normal colons, induced differentiation of tumor promoting inflammatory T helper 17 cells (Th17) cell responses in an IL-6 dependent manner. Our study suggests that CD90(+) fibroblasts/myofibroblasts may be the major source of IL-6 in T2-T3 CRC tumors, which supports the stemness of tumor cells and induces an immune adaptive inflammatory response (a.k.a. Th17) favoring tumor growth. Taken together our data supports the notion that IL-6 producing CAFs (a.k.a. C-CMFs) may provide a useful target for treating or preventing CRCs.
Collapse
Affiliation(s)
- Phuong T Huynh
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Ellen J Beswick
- Department of Molecular Genetics, University of New Mexico, Albuquerque, NM
| | - Yun A Coronado
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Paul Johnson
- Departments of Surgery, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Malaney R O'Connell
- Departments of Neuoroscience and Cell Biology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Tammara Watts
- Departments of Otolaryngology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Pomila Singh
- Departments of Neuoroscience and Cell Biology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Suimin Qiu
- Departments of Pathology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Katherine Morris
- Department of Surgery, University of New Mexico, Albuquerque, NM
| | - Don W Powell
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Departments of Neuoroscience and Cell Biology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| | - Irina V Pinchuk
- Departments of Internal Medicine, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Institute of Translational Science at University of Texas Medical Branch, Galveston, TX.,Departments of Microbiology and Immunology, Institute of Translational Science at University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
11
|
Role of intestinal myofibroblasts in HIV-associated intestinal collagen deposition and immune reconstitution following combination antiretroviral therapy. AIDS 2015; 29:877-88. [PMID: 25784439 DOI: 10.1097/qad.0000000000000636] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the potential role of mucosal intestinal myofibroblasts (IMFs) in HIV and associated fibrosis in gut-associated lymphoid tissue. DESIGN Profibrotic changes within the secondary lymphoid organs and mucosa have been implicated in failed immune reconstitution following effective combination antiretroviral therapy (cART). Microbial translocation is believed to be sustaining these systemic inflammatory pathways. IMFs are nonprofessional antigen-presenting cells with both immunoregulatory and mesenchymal functions that are ideally positioned to respond to translocating microbial antigen. METHODS Duodenal biopsies, obtained from patients naive to cART, underwent trichrome staining and were examined for tissue growth factor-beta (TGF-β) expression. Combined immunostaining and second harmonic generation analysis were used to determine IMF activation and collagen deposition. Confocal microscopy was performed to examine IMF activation and Toll-like receptor (TLR)4 expression. Finally, primary IMF cultures were stimulated with lipopolysaccharide to demonstrate the expression of the inflammatory biomarkers. RESULTS The expression of the fibrosis-promoting molecule, TGF-β1, is significantly increased in duodenal biopsies from HIV patients naïve to cART, and negatively correlated with subsequent peripheral CD4(+) recovery. The increase in TGF-β1 coincided with an increase in collagen deposition in the duodenal mucosa in the tissue area adjacent to the IMFs. We also observed that IMFs expressed TLR4 and had an activated phenotype since they were positive for fibroblast activation protein. Finally, stimulation of IMFs from HIV patients with TLR4 resulted in significantly increased expression of profibrotic molecules, TGF-β1, and interleukin-6. CONCLUSION Our data support the hypothesis that activated IMFs may be among the major cells contributing to the profibrotic changes, and thus, the establishment and maintenance of systemic inflammation interfering with immune reconstitution in HIV patients.
Collapse
|
12
|
Abstract
A growing body of evidence suggests that non-hematopoietic stromal cells of the intestine have multiple roles in immune responses and inflammation at this mucosal site. Despite this, many still consider gut stromal cells as passive structural entities, with past research focused heavily on their roles in fibrosis, tumor progression, and wound healing, rather than their contributions to immune function. In this review, we discuss our current knowledge of stromal cells in intestinal immunity, highlighting the many immunological axes in which stromal cells have a functional role. We also consider emerging data that broaden the potential scope of their contribution to immunity in the gut and argue that these so-called "non-immune" cells are reclassified in light of their diverse contributions to intestinal innate immunity and the maintenance of mucosal homeostasis.
Collapse
|
13
|
Cho JI, Joo IS, Choi JH, Jung KH, Choi EJ, Lee SH, Hwang IG. Prevalence and characterization of foodborne bacteria from meat products in Korea. Food Sci Biotechnol 2012. [DOI: 10.1007/s10068-012-0165-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
14
|
Okogbule-Wonodi AC, Li G, Anand B, Luzina IG, Atamas SP, Blanchard T. Human foetal intestinal fibroblasts are hyper-responsive to lipopolysaccharide stimulation. Dig Liver Dis 2012; 44:18-23. [PMID: 21956143 DOI: 10.1016/j.dld.2011.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 07/22/2011] [Accepted: 08/14/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND Intestinal myofibroblasts contribute to immune regulation in adults with inflammatory bowel disease but have not been characterised in neonatal intestinal inflammatory diseases. AIMS To compare lipopolysaccharide (LPS)-stimulated interleukin-8 (IL-8) production between human foetal and mature intestinal myofibroblasts in vitro. METHODS Foetal, neonatal and adult cells were stimulated with increasing concentrations of E. coli LPS. In LPS stimulated foetal myofibroblasts, Toll-like receptor 4 mRNA expression was assessed by real-time PCR whilst Toll-like receptor 4 receptor activity was determined using anti-Toll-like receptor 4 antibody. Mitogen activated protein kinase pathway activity was assessed using chemical inhibitors and Western blotting. IL-8 production was measured by quantitative ELISA. RESULTS IL-8 production by LPS stimulated foetal myofibroblasts occurred in a dose dependent manner. Toll-like receptor 4 expression was constitutive and Toll-like receptor 4 receptor blockade reduced IL-8 production by 42% (P=0.0262). C-Jun N-terminal kinase, p38 and NF-κB inhibitors significantly attenuated LPS stimulated IL-8 production by 42%, 33% and 2%, respectively. Mitogen activated protein kinase activity was confirmed by the presence of phosphorylated proteins on Western blots. CONCLUSION These data demonstrate increased IL-8 production by foetal myofibroblasts that is partially mediated by Toll-like receptor 4, mitogen activated protein kinase and NF-κB cell signalling pathways. Intestinal myofibroblasts cells may contribute to the dysregulated inflammatory response in the immature intestine and may form targets that lead to new therapies to prevent neonatal intestinal inflammatory bowel diseases.
Collapse
Affiliation(s)
- Adora C Okogbule-Wonodi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| | | | | | | | | | | |
Collapse
|
15
|
Hennekinne JA, De Buyser ML, Dragacci S. Staphylococcus aureus and its food poisoning toxins: characterization and outbreak investigation. FEMS Microbiol Rev 2011; 36:815-36. [PMID: 22091892 DOI: 10.1111/j.1574-6976.2011.00311.x] [Citation(s) in RCA: 563] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 09/15/2011] [Accepted: 10/03/2011] [Indexed: 11/29/2022] Open
Abstract
Staphylococcal food poisoning (SFP) is one of the most common food-borne diseases and results from the ingestion of staphylococcal enterotoxins (SEs) preformed in food by enterotoxigenic strains of Staphylococcus aureus. To date, more than 20 SEs have been described: SEA to SElV. All of them have superantigenic activity whereas half of them have been proved to be emetic, representing a potential hazard for consumers. This review, divided into four parts, will focus on the following: (1) the worldwide story of SFP outbreaks, (2) the characteristics and behaviour of S. aureus in food environment, (3) the toxinogenic conditions and characteristics of SEs, and (4) SFP outbreaks including symptomatology, occurrence in the European Union and currently available methods used to characterize staphylococcal outbreaks.
Collapse
Affiliation(s)
- Jacques-Antoine Hennekinne
- French Agency for Food, Environmental and Occupational Health & Safety (Anses), Food Safety Laboratory of Maisons-Alfort, European Union Reference Laboratory for Coagulase Positive Staphylococci, Maisons-Alfort, France.
| | | | | |
Collapse
|
16
|
Pinchuk IV, Beswick EJ, Saada JI, Boya G, Schmitt D, Raju GS, Brenmoehl J, Rogler G, Reyes VE, Powell DW. Human colonic myofibroblasts promote expansion of CD4+ CD25high Foxp3+ regulatory T cells. Gastroenterology 2011; 140:2019-30. [PMID: 21376048 PMCID: PMC3109194 DOI: 10.1053/j.gastro.2011.02.059] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 02/11/2011] [Accepted: 02/18/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Regulatory T (Treg) cells (CD4+ CD25high FoxP3+) regulate mucosal tolerance; their adoptive transfer prevents or reduces symptoms of colitis in mouse models of inflammatory bowel disease. Colonic CD90+ mesenchymal myofibroblasts and fibroblasts (CMFs) are abundant, nonprofessional antigen-presenting cells in the normal human colonic mucosa that suppress proliferation of activated CD4+ effector T cells. We studied CMF suppressive capacity and evaluated the ability of CMF to induce Treg cells. METHODS Allogeneic cocultures of CD4+ T cells and CMFs, derived from normal mucosa of patients undergoing colectomy for colon cancer or inflamed colonic tissues from patients with ulcerative colitis or Crohn's disease, were used to assess activation of the Treg cells. RESULTS Coculture of normal CMF with resting or naïve CD4+ T cells led to development of cells with a Treg phenotype; it also induced proliferation of a CD25+ CD127- FoxP3+ T cells, which expressed CTLA-4, interleukin-10, and transforming growth factor-β and had suppressive activities. In contrast to dendritic cells, normal CMFs required exogenous interleukin-2 to induce proliferation of naturally occurring Treg cells. Induction of Treg cells by normal CMFs required major histocompatibility complex class II and prostaglandin E2. CMFs from patients with inflammatory bowel diseases had reduced capacity to induce active Treg cells and increased capacity to transiently generate CD4+CD25+/- CD127+ T cells that express low levels of FoxP3. CONCLUSIONS CMFs suppress the immune response in normal colon tissue and might therefore help maintain colonic mucosal tolerance. Alterations in CMF-mediated induction of Treg cells might promote pathogenesis of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Irina V. Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Ellen J. Beswick
- Department of Molecular Genetics & Microbiology, University of New Mexico, Albuquerque, NM 87131
| | - Jamal I. Saada
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Gushyalatha Boya
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - David Schmitt
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas 77555
| | - Gottumukkala S. Raju
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Julia Brenmoehl
- Research Unit Genetics and Biometry, Leibnitz Institute of Farm Animal Biology Dummerstorf 18196, Germany
| | - Gerhard Rogler
- Departement für Innere Medizin, Klinik für Gastroenterologie und Hepatologie, Universitätsspital Zürich, Zürich, Schweiz
| | - Victor E. Reyes
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas 77555, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Don W. Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555, Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555,Corresponding author: Don W. Powell, M.D., University of Texas Medical Branch, 301, University Bld, Galveston, Tx 77555-0655, , Phone: (409) 772-1950 or 772-9015, Fax: (409) 772-8097 or 772-4789
| |
Collapse
|
17
|
Miron N, Miron MM. Staphylococcal enterotoxin A: a candidate for the amplification of physiological immunoregulatory responses in the gut. Microbiol Immunol 2011; 54:769-77. [PMID: 21091986 DOI: 10.1111/j.1348-0421.2010.00280.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Staphylococcal enterotoxin A (SEA) is one of the bacterial products tested for modulation of unwanted immune responses. Of all the staphylococcal enterotoxins, SEA is the most potent stimulator of T cells. When administered orally, SEA acts as a superantigen (SA), producing unspecific stimulation of intra-epithelial lymphocytes (IELs) in the intestinal mucosa. This stimulation results in amplification of the normal local immunologic responses, which are mainly regulatory. This amplification is based on increased local production of IFN-γ by IELs, which acts on the nearby enterocytes. As a result, the enterocytes produce large amounts of tolerosomes, cellular corpuscles which detach themselves from the basal poles of the enterocytes and contain antigenic peptides that are conditioned to be interpreted as tolerogenic by the gut immune system. Tolerosomes are physiologically produced as a response to dietary peptides; it is already known that enterocytes posses the molecular mechanisms for processing peptides in a similar manner to lymphocytes. The fate of tolerosomes is not precisely known, but it seems that they merge with intestinal dendritic cells, conveying to them the information that orally administered peptides must be interpreted as tolerogens. SEA can stimulate this mechanism, thus favoring the development of tolerance to peptides/proteins administered subsequently via the oral route. This characteristic of SEA might be useful in therapy for regulating immune responses. The present paper reviews the current status of research regarding the impact of SEA on the enteric immune system and its potential use in the treatment of allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Nicolae Miron
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj, Romania.
| | | |
Collapse
|
18
|
Abstract
The non-white blood cell mesenchymal elements of the intestinal lamina propria are the myofibroblasts, fibroblasts, pericytes, stromal stem cells, muscularis mucosae, and the smooth muscle of the villus core associated with the lymphatic lacteal. We review the functional anatomy of these mesenchymal cells, what is known about their origin in the embryo and their replacement in adults, their putative role in intestinal mucosal morphogenesis, and the intestinal stem cell niche, and we consider new information about myofibroblasts as nonprofessional immune cells. Although our knowledge of the function of mesenchymal cells in intestinal disease is rudimentary, we briefly consider here their roles in cancer and intestinal inflammation.
Collapse
Affiliation(s)
- I V Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555-0764, USA.
| | | | | | | |
Collapse
|
19
|
Abstract
The mesenchymal elements of the intestinal lamina propria reviewed here are the myofibroblasts, fibroblasts, mural cells (pericytes) of the vasculature, bone marrow-derived stromal stem cells, smooth muscle of the muscularis mucosae, and smooth muscle surrounding the lymphatic lacteals. These cells share similar marker molecules, origins, and coordinated biological functions previously ascribed solely to subepithelial myofibroblasts. We review the functional anatomy of intestinal mesenchymal cells and describe what is known about their origin in the embryo and their replacement in adults. As part of their putative role in intestinal mucosal morphogenesis, we consider the intestinal stem cell niche. Lastly, we review emerging information about myofibroblasts as nonprofessional immune cells that may be important as an alarm system for the gut and as a participant in peripheral immune tolerance.
Collapse
Affiliation(s)
- D.W. Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-0764
| | - I.V. Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
| | - J.I. Saada
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94143
| | - R.C. Mifflin
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555-0764
| |
Collapse
|
20
|
Abstract
Staphylococcus aureus (S. aureus) is a Gram positive bacterium that is carried by about one third of the general population and is responsible for common and serious diseases. These diseases include food poisoning and toxic shock syndrome, which are caused by exotoxins produced by S. aureus. Of the more than 20 Staphylococcal enterotoxins, SEA and SEB are the best characterized and are also regarded as superantigens because of their ability to bind to class II MHC molecules on antigen presenting cells and stimulate large populations of T cells that share variable regions on the β chain of the T cell receptor. The result of this massive T cell activation is a cytokine bolus leading to an acute toxic shock. These proteins are highly resistant to denaturation, which allows them to remain intact in contaminated food and trigger disease outbreaks. A recognized problem is the emergence of multi-drug resistant strains of S. aureus and these are a concern in the clinical setting as they are a common cause of antibiotic-associated diarrhea in hospitalized patients. In this review, we provide an overview of the current understanding of these proteins.
Collapse
|
21
|
Multiple roles of Staphylococcus aureus enterotoxins: pathogenicity, superantigenic activity, and correlation to antibiotic resistance. Toxins (Basel) 2010; 2:2117-31. [PMID: 22069676 PMCID: PMC3153285 DOI: 10.3390/toxins2082117] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Accepted: 08/09/2010] [Indexed: 12/03/2022] Open
Abstract
Heat-stable enterotoxins are the most notable virulence factors associated with Staphylococcus aureus, a common pathogen associated with serious community and hospital acquired diseases. Staphylococcal enterotoxins (SEs) cause toxic shock-like syndromes and have been implicated in food poisoning. But SEs also act as superantigens that stimulate T-cell proliferation, and a high correlation between these activities has been detected. Most of the nosocomial S. aureus infections are caused by methicillin-resistant S. aureus (MRSA) strains, and those resistant to quinolones or multiresistant to other antibiotics are emerging, leaving a limited choice for their control. This review focuses on these diverse roles of SE, their possible correlations and the influence in disease progression and therapy.
Collapse
|
22
|
Therapeutic down-modulators of staphylococcal superantigen-induced inflammation and toxic shock. Toxins (Basel) 2010; 2:1963-83. [PMID: 22069668 PMCID: PMC3153276 DOI: 10.3390/toxins2081963] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 07/16/2010] [Accepted: 07/28/2010] [Indexed: 12/21/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) and related superantigenic toxins are potent stimulators of the immune system and cause a variety of diseases in humans, ranging from food poisoning to toxic shock. These toxins bind directly to major histocompatibility complex (MHC) class II molecules on antigen-presenting cells and specific Vβ regions of T-cell receptors (TCR), resulting in hyperactivation of both monocytes/macrophages and T lymphocytes. Activated host cells produce massive amounts of proinflammatory cytokines and chemokines, activating inflammation and coagulation, causing clinical symptoms that include fever, hypotension, and shock. This review summarizes the in vitro and in vivo effects of staphylococcal superantigens, the role of pivotal mediators induced by these toxins in the pathogenic mechanisms of tissue injury, and the therapeutic agents to mitigate the toxic effects of superantigens.
Collapse
|
23
|
The systemic and pulmonary immune response to staphylococcal enterotoxins. Toxins (Basel) 2010; 2:1898-912. [PMID: 22069664 PMCID: PMC3153275 DOI: 10.3390/toxins2071898] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 07/12/2010] [Indexed: 11/21/2022] Open
Abstract
In response to environmental cues the human pathogen Staphylococcus aureus synthesizes and releases proteinaceous enterotoxins. These enterotoxins are natural etiologic entities of severe food poisoning, toxic shock syndrome, and acute diseases. Staphylococcal enterotoxins are currently listed as Category B Bioterrorism Agents by the Center for Disease Control and Prevention. They are associated with respiratory illnesses, and may contribute to exacerbation of pulmonary disease. This likely stems from the ability of Staphylococcal enterotoxins to elicit powerful episodes of T cell stimulation resulting in release of pro-inflammatory cytokines. Here, we discuss the role of the immune system and potential mechanisms of disease initiation and progression.
Collapse
|
24
|
Activation of adventitial fibroblasts in the early stage of the aortic transplant vasculopathy in rat. Transplantation 2010; 89:945-53. [PMID: 20098355 DOI: 10.1097/tp.0b013e3181d05aa7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Transplant vasculopathy (TV) is the most significant obstacle to long-term success of organ transplantation. Increasing attention has been paid to the role of adventitia in vascular diseases. We evaluated the role of adventitial fibroblasts in the development of TV. METHODS Thoracic aortas from Sprague-Dawley (SD) rats transplanted into the abdominal aortas of Wistar rats worked as allografts, and isografts (SD to SD) were control. Grafts were removed on days 3, 7, and 14 for histologic, morphometric, and immunohistochemical detection of vimentin, alpha-smooth muscle actin, Ki-67, CD3, transforming growth factor-beta1 (TGF-beta1), monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase-7 (MMP-7), and quantitative real-time reverse transcriptase polymerase chain reaction for TGF-beta1, MCP-1, MMP-7, tumor necrosis factor-alpha, and interleukin-1beta. RESULTS In the allografts, neointima thickness and neointima/media thickness ratios were slightly increased at 7 days and significantly increased at 14 days. Immunostaining of vimentin and alpha-smooth muscle actin showed adventitial fibroblasts activation and differentiation into myofibroblasts. Ki-67-positive nuclei were observed in the adventitia 3 days after allografting and subsequently in the neointima. No more than 4% CD3-positive cells were found in adventitia in all the groups. Compared with isografts, TGF-beta1, MMP-7, and MCP-1 were expressed in the adventitia before neointima formation and were significantly increased in allografts at all time points. Tumor necrosis factor-alpha and interleukin-1beta were also significantly increased in adventitia in allografts. CONCLUSIONS These results demonstrated that adventitial fibroblasts are activated and can produce cytokines and chemokines before the neointimal hyperplasia. They may exert a potential effect on the development of neointimal hyperplasia in TV.
Collapse
|
25
|
Dietz HC. TGF-beta in the pathogenesis and prevention of disease: a matter of aneurysmic proportions. J Clin Invest 2010; 120:403-7. [PMID: 20101091 DOI: 10.1172/jci42014] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
TGF-beta regulates many aspects of cellular performance relevant to tissue morphogenesis and homeostasis. Postnatal perturbation of TGF-beta signaling contributes to the pathogenesis of many disease states, as recently exemplified through the study of Marfan syndrome (MFS), including aortic aneurysm and skeletal muscle myopathy. Heterogeneity in the regulation and consequences of TGF-beta signaling, amplified in the context of disease, has engendered confusion and controversy regarding its utility as a therapeutic target. Three studies recently published in the JCI, including one in this issue, underscore the complexity of this subject. Heydemann and colleagues implicate dimorphic variation in latent TGF-beta-binding protein 4 (LTBP4), a regulator of TGF-beta bioavailability and activation, as a modifier of muscular dystrophy in gamma-sarcoglycan-deficient mice. In contrast to experience with ascending aortic aneurysm in MFS, Wang and colleagues show that systemic abrogation of TGF-beta signaling worsens (rather than attenuates) Ang II-induced abdominal aortic aneurysm progression in mice. Tieu and colleagues define alterations in the regulation of vascular inflammation in the pathogenesis of Ang II-induced aneurysm and dissection in mice, which may help shed some light on this apparent paradox.
Collapse
Affiliation(s)
- Harry C Dietz
- Institute of Genetic Medicine and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| |
Collapse
|
26
|
Francoeur C, Bouatrouss Y, Seltana A, Pinchuk IV, Vachon PH, Powell DW, Sawan B, Seidman EG, Beaulieu JF. Degeneration of the pericryptal myofibroblast sheath by proinflammatory cytokines in inflammatory bowel diseases. Gastroenterology 2009; 136:268-277.e3. [PMID: 18976656 DOI: 10.1053/j.gastro.2008.10.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 09/27/2008] [Accepted: 10/02/2008] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS Inflammatory bowel diseases (IBDs) are characterized by remodeling of the intestinal mucosa, which is associated with excessive cytokine release. Previous studies have shown that the epithelium in the crypt region of the mucosa in patients with Crohn's disease is susceptible to proinflammatory cytokines. We investigated whether the subepithelial myofibroblasts in this region were affected by these inflammatory conditions. METHODS Immunofluorescence and immunohistochemistry were performed on inflamed and uninflamed specimens from patients with IBD to detect alpha-smooth muscle actin (alphaSMA), desmin, and tenascin-C. The effects of the proinflammatory cytokines interleukin-1beta, tumor necrosis factor-alpha, and interferon-gamma were analyzed in human intestinal myofibroblast cultures by immunoblotting and apoptosis assays. RESULTS Immunofluorescence analysis revealed decreased levels of the extracellular matrix molecule tenascin-C in pericryptal sheaths and alphaSMA in the immediate vicinity of the crypts in the inflamed specimens, indicating that the myofibroblast pericryptal sheath is affected by proinflammatory cytokines. Although individual cytokines did not affect myofibroblast proliferation or survival, cytokine combinations triggered caspase-dependent apoptosis. alphaSMA levels were reduced significantly in cells exposed to cytokines, either alone or in combination, suggesting dedifferentiation of myofibroblasts. Proinflammatory cytokines did not affect tenascin-C expression, suggesting that the decrease observed in the inflamed mucosa resulted from myofibroblast apoptosis. CONCLUSIONS The subepithelial myofibroblasts of the epithelial sheath are disrupted in the intestinal mucosa of patients with IBD. A loss of myofibroblasts appears to result from the susceptibility of these cells to proinflammatory cytokines.
Collapse
Affiliation(s)
- Caroline Francoeur
- Canadian Institutes of Health Research Team on the Digestive Epithelium, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Pinchuk IV, Saada JI, Beswick EJ, Boya G, Qiu SM, Mifflin RC, Raju GS, Reyes VE, Powell DW. PD-1 ligand expression by human colonic myofibroblasts/fibroblasts regulates CD4+ T-cell activity. Gastroenterology 2008; 135:1228-1237, 1237.e1-2. [PMID: 18760278 PMCID: PMC2584612 DOI: 10.1053/j.gastro.2008.07.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 06/13/2008] [Accepted: 07/03/2008] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS A prominent role for inhibitory molecules PD-L1 and PD-L2 in peripheral tolerance has been proposed. However, the phenotype and function of PD-L-expressing cells in human gut remains unclear. Recent studies suggest that colonic myofibroblasts (CMFs) and fibroblasts are important in the switch from acute inflammation to adaptive immunity. In the normal human colon, CMFs represent a distinct population of major histocompatibility complex class II(+) cells involved in the regulation of mucosal CD4(+) T-cell responses. METHODS PD-L1 and PD-L2 expression on human CMFs was determined using Western blot, fluorescence-activated cell sorter analysis and confocal microscopy. Lymphoproliferation assays and cytokine enzyme-linked immunosorbent assays were used to evaluate the role of B7 costimulators expressed by CMFs with regard to the regulation of preactivated T-helper cell responses. RESULTS We demonstrate here the expression of PD-L1/2 molecules by normal human CMF and fibroblasts in situ and in culture. Both molecules support suppressive functions of CMFs in the regulation of activated CD4(+) T-helper cell proliferative responses; blocking this interaction reverses the suppressive effect of CMFs on T-cell proliferation and leads to increased production of the major T-cell growth factor, interleukin (IL)-2. PD-L1/2-mediated CMF suppressive functions are mainly due to the inhibition of IL-2 production, because supplementation of the coculture media with exogenous IL-2 led to partial recovery of activated T-cell proliferation. CONCLUSIONS Our data suggest that stromal myofibroblasts and fibroblasts may limit T-helper cell proliferative activity in the gut and, thus, might play a prominent role in mucosal intestinal tolerance.
Collapse
Affiliation(s)
- Irina V. Pinchuk
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas 77555
| | - Jamal I. Saada
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Ellen J. Beswick
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas 77555
| | - Gushyalatha Boya
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Sumin M. Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Randy C. Mifflin
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Gottumukkala S. Raju
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Victor E. Reyes
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas 77555
| | - Don W. Powell
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555, Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555
| |
Collapse
|