1
|
Shen Y, Gong Z, Zhang S, Cao J, Mao W, Yao Y, Zhao J, Li Q, Liu K, Liu B, Feng S. Besides TLR2 and TLR4, NLRP3 is also involved in regulating Escherichia coli infection-induced inflammatory responses in mice. Int Immunopharmacol 2023; 121:110556. [PMID: 37364329 DOI: 10.1016/j.intimp.2023.110556] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
The host Toll-like Receptor-2 (TLR2) and Toll-like Receptor-4 (TLR4) play critical roles in defense against Escherichia coli (E. coli) infection is well-known. The NLR pyrin domain-containing 3 (NLRP3) inflammasome is also an important candidate during the host-recognized pathogen, while the roles of NLRP3 in the host inflammatory response to E. coli infection remains unclear. This study aimed to explore the roles of NLRP3 in regulating the inflammatory response in E. coli infection-induced mice. Our result indicated that compared to wild-type mice, the TLR2-deficient (TLR2-/-), TLR4-deficient (TLR4-/-), and NLRP3-deficient (NLRP3-/-) mice had significant decrease in liver damage after stimulation with Lipopolysaccharide (LPS, 1 μg/mL), Braun lipoprotein (BLP, 1 μg/mL), or infected by WT E. coli (1 × 107 CFU, MOI 5:1). Meanwhile, compared with wild-type mice, the TNF-α and IL-1β production in serum decreased in TLR2-/-, TLR4-/-, and NLRP3-/- mice after LPS, BLP treatment, or WT E. coli infection. In macrophages from NLRP3-/- mice showed significantly reduced secretion of TNF-α and IL-1β in response to stimulation with LPS, BLP, or WT E. coli infection compared with macrophages from wild-type mice. These results indicate that besides TLR2 and TLR4, NLRP3 also plays a critical role in host inflammatory responses to defense against E. coli infection, and might provide a therapeutic target in combating disease with bacterium infection.
Collapse
Affiliation(s)
- Yuan Shen
- Key Laboratory of Molecular Epidemiology of Chronic Diseases, School of Public Health, Inner Mongolia Medical University, No. 5, Xinhua Street, Hui Min District, 010000, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Zhiguo Gong
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Shuangyi Zhang
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Jinshan Cao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Wei Mao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Yuan Yao
- Department of Neurology, Inner Mongolia People's Hospital, No. 20, Zhaowuda Road, Saihan District, 010017, Hohhot City, China
| | - Jiamin Zhao
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Qianru Li
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Kun Liu
- Key Laboratory of Molecular Epidemiology of Chronic Diseases, School of Public Health, Inner Mongolia Medical University, No. 5, Xinhua Street, Hui Min District, 010000, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China
| | - Bo Liu
- Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China.
| | - Shuang Feng
- Laboratory of Veterinary Public Health, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot City, China.
| |
Collapse
|
2
|
Alshaikh BN, Reyes Loredo A, Knauff M, Momin S, Moossavi S. The Role of Dietary Fats in the Development and Prevention of Necrotizing Enterocolitis. Nutrients 2021; 14:145. [PMID: 35011027 PMCID: PMC8746672 DOI: 10.3390/nu14010145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 11/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a significant cause of mortality and morbidity in preterm infants. The pathogenesis of NEC is not completely understood; however, intestinal immaturity and excessive immunoreactivity of intestinal mucosa to intraluminal microbes and nutrients appear to have critical roles. Dietary fats are not only the main source of energy for preterm infants, but also exert potent effects on intestinal development, intestinal microbial colonization, immune function, and inflammatory response. Preterm infants have a relatively low capacity to digest and absorb triglyceride fat. Fat may thereby accumulate in the ileum and contribute to the development of NEC by inducing oxidative stress and inflammation. Some fat components, such as long-chain polyunsaturated fatty acids (LC-PUFAs), also exert immunomodulatory roles during the early postnatal period when the immune system is rapidly developing. LC-PUFAs may have the ability to modulate the inflammatory process of NEC, particularly when the balance between n3 and n6 LC-PUFAs derivatives is maintained. Supplementation with n3 LC-PUFAs alone may have limited effect on NEC prevention. In this review, we describe how various fatty acids play different roles in the pathogenesis of NEC in preterm infants.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Adriana Reyes Loredo
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Megan Knauff
- Nutrition Services, Alberta Health Services, Calgary, AB T2N 2T9, Canada
| | - Sarfaraz Momin
- Neonatal Nutrition and Gastroenterology Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
| | - Shirin Moossavi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
- International Microbiome Centre, Cumming School of Medicine, Health Sciences Centre, University of Calgary, Calgary, AB T2N 2T9, Canada
| |
Collapse
|
3
|
Sampah MES, Hackam DJ. Prenatal Immunity and Influences on Necrotizing Enterocolitis and Associated Neonatal Disorders. Front Immunol 2021; 12:650709. [PMID: 33968047 PMCID: PMC8097145 DOI: 10.3389/fimmu.2021.650709] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Prior to birth, the neonate has limited exposure to pathogens. The transition from the intra-uterine to the postnatal environment initiates a series of complex interactions between the newborn host and a variety of potential pathogens that persist over the first few weeks of life. This transition is particularly complex in the case of the premature and very low birth weight infant, who may be susceptible to many disorders as a result of an immature and underdeveloped immune system. Chief amongst these disorders is necrotizing enterocolitis (NEC), an acute inflammatory disorder that leads to necrosis of the intestine, and which can affect multiple systems and have the potential to result in long term effects if the infant is to survive. Here, we examine what is known about the interplay of the immune system with the maternal uterine environment, microbes, nutritional and other factors in the pathogenesis of neonatal pathologies such as NEC, while also taking into consideration the effects on the long-term health of affected children.
Collapse
Affiliation(s)
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine , Baltimore, MD, United States
| |
Collapse
|
4
|
Trauma-induced lung injury is associated with infiltration of activated TLR expressing myeloid cells. Cytokine 2021; 141:155457. [PMID: 33581471 DOI: 10.1016/j.cyto.2021.155457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Traumatic injury with hemorrhage (TH) induces an inflammatory response in the lung resulting in lung injury involving activation of immune cells including myeloid cells (i.e., monocytes, granulocytes and macrophages), in part through TLRs. TLRs, via the recognition of damage associated molecular patterns (DAMPs), are a key link between tissue injury and inflammation. Nonetheless, the role of TLRs in myeloid cell activation and TH-induced lung injury remains ill defined. METHODS C57BL/6 male mice were subjected to TH or sham treatment (n = 4-6 /group). Lung tissues were collected two hrs. after injury. Single cells were isolated from the lungs by enzymatic digestion and myeloid cell TLR expression and activation (i.e., cytokine production) were assessed using flow cytometry techniques. RESULTS The injury was associated with a profound change in the lung myeloid cell population. TH markedly increased lung CD11b+ monocyte numbers and Gr1+ granulocyte numbers as compared to sham mice. The number of cells expressing TLR2, TLR4, and TLR9 were increased 4-7 fold in the TH mice. Activation for elevated cytokine (TNFα, IL-10) production was observed in the lung monocyte population of the TH mice. CONCLUSIONS Trauma-induced lung injury is associated with infiltration of the lungs with TLR expressing myeloid cells that are activated for elevated cytokine responses. This elevation in TLR expression may contribute to DAMP-mediated pulmonary complications of an inflammatory nature and warrants further investigation.
Collapse
|
5
|
Stephens M, von der Weid PY. Lipopolysaccharides modulate intestinal epithelial permeability and inflammation in a species-specific manner. Gut Microbes 2020; 11:421-432. [PMID: 31203717 PMCID: PMC7524286 DOI: 10.1080/19490976.2019.1629235] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Patients presenting with Inflammatory bowel disease have been shown to exhibit an altered microbiome in both Crohn's disease and Ulcerative colitis. This shift in the microbial content led us to question whether several of these microbes are important in inflammatory processes present in these diseases and more specifically whether lipopolysaccharides from the gram-negative cell wall differentially stimulates resident cells. We, therefore, investigated the possible contribution of five major species of gram-negative bacteria found to be altered in presence during disease progression and evaluate their pathogenicity through LPS. We demonstrated that LPS from these different species had individual capacities to induce NF-κB and pro-inflammatory IL-8 production from HEK-TLR4 cells in a TLR4 dependent manner. Additional work using human intestinal colonic epithelial cell monolayers (Caco-2) demonstrated that the cells responded to the serotype specific LPS in a distinct manner, inducing many inflammatory mediators such as TNF-α and IL-10 in significantly altered proportions. Furthermore, the permeability of Caco-2 monolayers, as a test for their ability to alter intestinal permeability, was also differentially altered by the serotype specific LPS modulating trans-epithelial electrical resistance, small molecule movement, and tight junction integrity. Our results suggest that specific species of bacteria may be potentiating the pathogenesis of IBD and chronic inflammatory diseases through their serotype specific LPS responses.
Collapse
Affiliation(s)
- Matthew Stephens
- Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Pierre-Yves von der Weid
- Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada,CONTACT Pierre-Yves von der Weid Snyder Institute of Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
6
|
Kim CS, Claud EC. Necrotizing Enterocolitis Pathophysiology: How Microbiome Data Alter Our Understanding. Clin Perinatol 2019; 46:29-38. [PMID: 30771817 PMCID: PMC6816463 DOI: 10.1016/j.clp.2018.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Necrotizing enterocolitis is a major cause of mortality and morbidity in the preterm infant population. The gut microbiome is of particular interest in research surrounding necrotizing enterocolitis, because variations in the intestinal microbiota seem to correlate with the risk of inflammation and disease. Recent advances in non-culture-based genomic sequencing have also allowed for more intricate analyses of the intestinal microbiome. Its evolution seems to be influenced by intrauterine and extrauterine factors, ranging from antenatal antibiotic exposure to type of enteral feeds. Ultimately, these alterations in the gut microbiome have the potential to result in devastating diseases like necrotizing enterocolitis.
Collapse
Affiliation(s)
- Christina S. Kim
- Neonatology, Department of Pediatrics, University of Chicago, Chicago, Illinois, United States
| | - Erika C. Claud
- Neonatology, Department of Pediatrics, University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
7
|
Denning NL, Prince JM. Neonatal intestinal dysbiosis in necrotizing enterocolitis. Mol Med 2018; 24:4. [PMID: 30134786 PMCID: PMC6016883 DOI: 10.1186/s10020-018-0002-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022] Open
Abstract
Necrotizing Enterocolitis (NEC) is one of the most devastating gastrointestinal diseases in neonates, particularly among preterm infants in whom surgical NEC is the leading cause of morbidity. NEC pathophysiology occurs in the hyper-reactive milieu of the premature gut after bacterial colonization. The resultant activation of the TLR4 pathway appears to be a strongly contributing factor. Advancements in metagenomics may yield new clarity to the relationship between the neonatal intestinal microbiome and the development of NEC. After a century without effective directed treatments, microbiome manipulation offers a promising therapeutic target for the prevention and treatment of this devastating disease.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA. .,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.
| | - Jose M Prince
- Division of Pediatric Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, 269-01 76th Avenue, CH 158, New Hyde Park, New York, NY, 11040, USA.,Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.,Trauma Institute, Northwell Health System, Manhasset, NY, 11030, USA
| |
Collapse
|
8
|
Cukrowska B, Sowińska A, Bierła JB, Czarnowska E, Rybak A, Grzybowska-Chlebowczyk U. Intestinal epithelium, intraepithelial lymphocytes and the gut microbiota - Key players in the pathogenesis of celiac disease. World J Gastroenterol 2017; 23:7505-7518. [PMID: 29204051 PMCID: PMC5698244 DOI: 10.3748/wjg.v23.i42.7505] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/31/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023] Open
Abstract
Celiac disease (CD) is a chronic immune-mediated disorder triggered by the ingestion of gluten in genetically predisposed individuals. Before activating the immune system, gluten peptides are transferred by the epithelial barrier to the mucosal lamina propria, where they are deamidated by intestinal tissue transglutaminase 2. As a result, they strongly bind to human leucocyte antigens (HLAs), especially HLA-DQ2 and HLA-DQ8, expressed on antigen-presenting cells. This induces an inflammatory response, which results in small bowel enteropathy. Although gluten is the main external trigger activating both innate and adaptive (specific) immunity, its presence in the intestinal lumen does not fully explain CD pathogenesis. It has been hypothesized that an early disruption of the gut barrier in genetically susceptible individuals, which would result in an increased intestinal permeability, could precede the onset of gluten-induced immune events. The intestinal barrier is a complex functional structure, whose functioning is dependent on intestinal microbiota homeostasis, epithelial layer integrity, and the gut-associated lymphoid tissue with its intraepithelial lymphocytes (IELs). The aim of this paper was to review the current literature and summarize the role of the gut microbiota, epithelial cells and their intercellular junctions, and IELs in CD development.
Collapse
Affiliation(s)
- Bożena Cukrowska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Agnieszka Sowińska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Joanna Beata Bierła
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Anna Rybak
- Department of Gastroenterology, Division of Neurogastroenterology and Motility, Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | | |
Collapse
|
9
|
Mett V, Komarova EA, Greene K, Bespalov I, Brackett C, Gillard B, Gleiberman AS, Toshkov IA, Aygün-Sunar S, Johnson C, Karasik E, Bapardekar-Nair M, Kurnasov OV, Osterman AL, Stanhope-Baker PS, Morrison C, Moser MT, Foster BA, Gudkov AV. Mobilan: a recombinant adenovirus carrying Toll-like receptor 5 self-activating cassette for cancer immunotherapy. Oncogene 2017; 37:439-449. [PMID: 28967901 PMCID: PMC5799711 DOI: 10.1038/onc.2017.346] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/23/2017] [Accepted: 08/07/2017] [Indexed: 12/14/2022]
Abstract
Toll-like receptor 5 (TLR5) is considered an attractive target for anticancer immunotherapy. TLR5 agonists, bacterial flagellin and engineered flagellin derivatives, have been shown to have potent antitumor and metastasis-suppressive effects in multiple animal models and to be safe in both animals and humans. Anticancer efficacy of TLR5 agonists stems from TLR5-dependent activation of nuclear factor-κB (NF-κB) that mediates innate and adaptive antitumor immune responses. To extend application of TLR5-targeted anticancer immunotherapy to tumors that do not naturally express TLR5, we created an adenovirus-based vector for intratumor delivery, named Mobilan that drives expression of self-activating TLR5 signaling cassette comprising of human TLR5 and a secreted derivative of Salmonella flagellin structurally analogous to a clinical stage TLR5 agonist, entolimod. Co-expression of TLR5 receptor and agonist in Mobilan-infected cells established an autocrine/paracrine TLR5 signaling loop resulting in constitutive activation of NF-κB both in vitro and in vivo. Injection of Mobilan into primary tumors of the prostate cancer-prone transgenic adenocarcinoma of the mouse prostate (TRAMP) mice resulted in a strong induction of multiple genes involved in inflammatory responses and mobilization of innate immune cells into the tumors including neutrophils and NK cells and suppressed tumor progression. Intratumoral injection of Mobilan into subcutaneously growing syngeneic prostate tumors in immunocompetent hosts improved animal survival after surgical resection of the tumors, by suppression of tumor metastasis. In addition, vaccination of mice with irradiated Mobilan-transduced prostate tumor cells protected mice against subsequent tumor challenge. These results provide proof-of-concept for Mobilan as a tool for antitumor vaccination that directs TLR5-mediated immune response toward cancer cells and does not require identification of tumor antigens.
Collapse
Affiliation(s)
- V Mett
- Cleveland Biolabs Inc., Buffalo, NY, USA
| | - E A Komarova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K Greene
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - I Bespalov
- Cleveland Biolabs Inc., Buffalo, NY, USA
| | - C Brackett
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - B Gillard
- Department of Molecular Pharmacology and Cancer Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | | | | - C Johnson
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - E Karasik
- Department of Molecular Pharmacology and Cancer Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - O V Kurnasov
- Infectious and Inflammatory Disease Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - A L Osterman
- Infectious and Inflammatory Disease Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - C Morrison
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - M T Moser
- Department of Molecular Pharmacology and Cancer Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - B A Foster
- Department of Molecular Pharmacology and Cancer Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - A V Gudkov
- Cleveland Biolabs Inc., Buffalo, NY, USA.,Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
10
|
Pierzchalska M, Panek M, Czyrnek M, Gielicz A, Mickowska B, Grabacka M. Probiotic Lactobacillus acidophilus bacteria or synthetic TLR2 agonist boost the growth of chicken embryo intestinal organoids in cultures comprising epithelial cells and myofibroblasts. Comp Immunol Microbiol Infect Dis 2017; 53:7-18. [PMID: 28750869 DOI: 10.1016/j.cimid.2017.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 05/31/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023]
Abstract
The intestinal epithelial cells reside in close proximity to myofibroblasts and microbiota, which are supposed to have an impact on intestinal stem cells fate and to influence processes of tissue maturation and regeneration. Mechanism underlying these phenomena and their diversity among vertebrates can be studied in 3D organoid cultures. We investigated the growth of chicken embryo intestinal epithelial organoids in Matrigel with and without Toll-like receptors (TLRs) stimulation. The organoid cultures contained also some myofibroblasts with potential to promote intestinal stem cell survival. Organoid cells, expressing TLR4, TLR2 type 1 and TLR2 type 2 were incubated with their agonists (lipopolysaccharide - LPS and Pam3CSK4) or co-cultured with Lactobacillus acidophilus bacteria (LA-5). Pam3CSK4 and LA-5 promoted organoid growth, which was demonstrated by comparing the morphological parameters (mean number and area of organoids). The profile of prostaglandins (PG), known to promote intestinal regeneration, in supernatants from organoid and fibroblast cultures were evaluated. Both PGE2 and PGD2 were detected. As compared to unstimulated controls, supernatants from the Pam3CSK4-stimulated organoids contained twice as much of PGE2 and PGD2. The changes in production of prostaglandins and the support of epithelial cell growth by myofibroblasts are factors potentially responsible for stimulatory effect of TLR2 activation.
Collapse
Affiliation(s)
- Malgorzata Pierzchalska
- Department of Food Biotechnology, Faculty of Food Technology, The University of Agriculture in Kraków, Balicka 122, 30-149, Kraków, Poland.
| | - Malgorzata Panek
- Department of Food Biotechnology, Faculty of Food Technology, The University of Agriculture in Kraków, Balicka 122, 30-149, Kraków, Poland
| | - Malgorzata Czyrnek
- Department of Food Biotechnology, Faculty of Food Technology, The University of Agriculture in Kraków, Balicka 122, 30-149, Kraków, Poland
| | - Anna Gielicz
- Department of Medicine, Jagiellonian University Medical College, Skawinska 8, 31-066 Kraków, Poland
| | - Barbara Mickowska
- Malopolska Center of Food Monitoring, Faculty of Food Technology, The University of Agriculture in Kraków, Balicka 122, 30-149, Kraków, Poland
| | - Maja Grabacka
- Department of Food Biotechnology, Faculty of Food Technology, The University of Agriculture in Kraków, Balicka 122, 30-149, Kraków, Poland
| |
Collapse
|
11
|
MyD88 gene knockout attenuates paraquat-induced acute lung injury. Toxicol Lett 2017; 269:41-46. [DOI: 10.1016/j.toxlet.2017.01.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/17/2017] [Accepted: 01/21/2017] [Indexed: 12/19/2022]
|
12
|
Yin Y, Liu F, Li Y, Tang R, Wang J. mRNA expression of TLR4, TLR9 and NF-κB in a neonatal murine model of necrotizing enterocolitis. Mol Med Rep 2016; 14:1953-6. [PMID: 27357505 PMCID: PMC4991741 DOI: 10.3892/mmr.2016.5455] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022] Open
Abstract
A neonatal model of necrotizing enterocolitis (NEC) in mice was established to examine the role of Toll-like receptors (TLRs) 4 and 9, and of nuclear factor (NF)-κB by quantitative detection of their mRNAs in intestinal tissue during the occurrence of NEC, and thus aid in the understanding of the basic pathogenesis of NEC. A total of 50 newborn BALB/c mice (specific pathogen-free level) ranging in age from 7 to 10 days, of either gender, and weighing 4.8–5.4 g were selected and randomly divided into a control and test group, n=25 mice per group. Mice in the control group were kept in the same cage with the mother who fed them, free from any interventions. Mice in the test group were separated from their mother 48 h following birth and placed in an incubator, artificially fed with milk substitutes, and regularly treated with hypoxia and cold stimulation (100% nitrogen anoxia for 90 sec, cold stimulation at 4°C for 10 min, 3 times a day for 3 days) to induce the neonatal NEC. The general state and body weight variations of the mice were recorded, the mice were sacrificed and the intestinal tissue necrosis was evaluated visually, the degree of intestinal injury was determined by histopathological staining, and the mRNA expression levels of intestinal tissue TLR4, TLR9 and NF-κB were quantified. Of the 25 mice in the test group, 3 died a natural death and 22 were sacrificed; their general state was worse than that of the mice in the control group, and the body weight variations among them were considerably larger. NEC was confirmed in 12 cases by visual inspection, and the average histological scores of the mice in the test group were 3.5±0.6, significantly higher than that in the control group (P<0.05). The mRNA expression of TLR4 and NF-κB in the test group were significantly higher than in the control group. By contrast, the mRNA expression of TLR9 was significantly lower in the test group, and differences were statistically significant (P<0.05). Thus, the increased mRNA expression of TLR4 and NF-κB, and decreased mRNA expression of TLR9 during NEC may be an important inflammatory mechanism of the disease.
Collapse
Affiliation(s)
- Yiyu Yin
- Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Fengli Liu
- Xuzhou Children's Hospital, Xuzhou, Jiangsu 221006, P.R. China
| | - Yiping Li
- Pediatric Research Institute of Soochow University. Suzhou, Jiangsu 215025, P.R. China
| | - Ruze Tang
- Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| | - Jian Wang
- Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, P.R. China
| |
Collapse
|
13
|
Qi W, Shen Q, Zhang L, Han LP, Wang S. Study on the inflammatory intervention of erythropoietin on NEC. Exp Ther Med 2016; 11:2221-2224. [PMID: 27284304 PMCID: PMC4887799 DOI: 10.3892/etm.2016.3199] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/29/2016] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate the effect of erythropoietin (EPO) on the inflammatory response and the mechanism analysis of the Τoll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling pathway of NEC. A total of 94 patients with necrotizing enterocolitis (NEC) were randomly divided into the control (42 cases) and observation (52 cases) groups, The control group received the standard medical treatment plan, whereas for the observation group this treatment plan was combined with the application of recombinant EPO for intramuscular injection treatment. The clinical effect was subsequently compared. The results showed that the complication and death rates in the observation group were significantly lower than those in the control group with statistically significant differences (P<0.05). Following treatments, the levels of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 in the observation group were significantly lower than those in the control group. The expression levels of mRNA of TLR4 and NF-κB in the observation group were significantly lower than those in the control group, with statistically significant differences (P<0.05). In summary, EPO was able to reduce the levels of inflammatory response of TNF-α and IL-6 through the TLR4/NF-κB signaling pathway, and improve the NEC, thus providing a basis for the clinical treatment of NEC.
Collapse
Affiliation(s)
- Weibin Qi
- Department of Neonatology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Qiong Shen
- Department of Gynaecology and Obstetrics, Armed Police Hebei Corps Hospital, Shijiazhuang, Hebei 050081, P.R. China
| | - Lin Zhang
- Department of Pediatrics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Li-Ping Han
- Department of Neonatology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Sumin Wang
- Department of Neonatology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
14
|
Kaplan BLF, Li J, LaPres JJ, Pruett SB, Karmaus PWF. Contributions of nonhematopoietic cells and mediators to immune responses: implications for immunotoxicology. Toxicol Sci 2016; 145:214-32. [PMID: 26008184 DOI: 10.1093/toxsci/kfv060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunotoxicology assessments have historically focused on the effects that xenobiotics exhibit directly on immune cells. These studies are invaluable as they identify immune cell targets and help characterize mechanisms and/or adverse outcome pathways of xenobiotics within the immune system. However, leukocytes can receive environmental cues by cell-cell contact or via released mediators from cells of organs outside of the immune system. These organs include, but are not limited to, the mucosal areas such as the lung and the gut, the liver, and the central nervous system. Homeostatic perturbation in these organs induced directly by toxicants can initiate and alter the outcome of local and systemic immunity. This review will highlight some of the identified nonimmune influences on immune homeostasis and provide summaries of how immunotoxic mechanisms of selected xenobiotics involve nonimmune cells or mediators. Thus, this review will identify data gaps and provide possible alternative mechanisms by which xenobiotics alter immune function that could be considered during immunotoxicology safety assessment.
Collapse
Affiliation(s)
- Barbara L F Kaplan
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Jinze Li
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - John J LaPres
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Stephen B Pruett
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Peer W F Karmaus
- *Center for Environmental Health Sciences, Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi St, Mississippi 39762, Safety Assessment, Genentech, Inc. South San Francisco, California 94080, Department of Biochemistry and Molecular Biology, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824 and Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| |
Collapse
|
15
|
The Influence of the Gut Microbiome on Obesity, Metabolic Syndrome and Gastrointestinal Disease. Clin Transl Gastroenterol 2015; 6:e91. [PMID: 26087059 PMCID: PMC4816244 DOI: 10.1038/ctg.2015.16] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/13/2015] [Indexed: 12/11/2022] Open
Abstract
There is a fine balance in the mutual relationship between the intestinal microbiota and its mammalian host. It is thought that disruptions in this fine balance contribute/account for the pathogenesis of many diseases. Recently, the significance of the relationship between gut microbiota and its mammalian host in the pathogenesis of obesity and the metabolic syndrome has been demonstrated. Emerging data has linked intestinal dysbiosis to several gastrointestinal diseases including inflammatory bowel disease, irritable bowel syndrome, nonalcoholic fatty liver disease, and gastrointestinal malignancy. This article is intended to review the role of gut microbiota maintenance/alterations of gut microbiota as a significant factor as a significant factor discriminating between health and common diseases. Based on current available data, the role of microbial manipulation in disease management remains to be further defined and a focus for further clinical investigation.
Collapse
|
16
|
Lee SC, Hsu JS, Li CC, Chen KM, Liu CT. Protective effect of leaf essential oil from Cinnamomum osmophloeum Kanehira on endotoxin-induced intestinal injury in mice associated with suppressed local expression of molecules in the signaling pathways of TLR4 and NLRP3. PLoS One 2015; 10:e0120700. [PMID: 25794175 PMCID: PMC4368640 DOI: 10.1371/journal.pone.0120700] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/26/2015] [Indexed: 02/07/2023] Open
Abstract
Endotoxin is a potent microbial mediator implicated in sepsis. We investigated the anti-inflammatory effect of leaf essential oil from Cinnamomum osmophloeum Kanehira (CO) of the linalool chemotype on endotoxin-injected mice. Mice were administered CO or vehicle by gavage before endotoxin injection and were killed 12 h after injection. Neither growth nor the organ weight or tissue weight to body weight ratio was affected by CO treatment. CO significantly lowered peripheral levels of tumor necrosis factor-α, interleukin (IL)-1β, IL-18, interferon-γ, and nitric oxide and inhibited the expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response gene (88), myeloid differentiation factor 2, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), caspase-1, and Nod-like receptor family, pyrin domain containing 3 (NLRP3). CO also inhibited the activation of nuclear factor-ĸB, inhibited the activity of caspase-1 in small intestine, and ameliorated intestinal edema. Our data provide strong evidence for a protective effect of CO of the linalool chemotype in the endotoxin-induced systemic inflammatory response in close association with suppression of the TLR4 and NLRP3 signaling pathways in intestine.
Collapse
Affiliation(s)
- Shih-Chieh Lee
- Department of BioIndustry Technology, Da-Yeh University, No. 168, University Rd., Dacun, Changhua, Taiwan, Republic of China
| | - Jie-Sheng Hsu
- School of Nutrition, Chung Shan Medical University, No. 110, Sec. 1, Chien Kuo N. Rd., Taichung, Taiwan, Republic of China
| | - Chien-Chun Li
- School of Nutrition, Chung Shan Medical University, No. 110, Sec. 1, Chien Kuo N. Rd., Taichung, Taiwan, Republic of China
| | - Ke-Ming Chen
- Department of Parasitology, Chung Shan Medical University, No. 110, Sec. 1, Chien Kuo N. Rd., Taichung, Taiwan, Republic of China
| | - Cheng-Tzu Liu
- School of Nutrition, Chung Shan Medical University, No. 110, Sec. 1, Chien Kuo N. Rd., Taichung, Taiwan, Republic of China
- Department of Nutrition, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien Kuo N. Rd., Taichung, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
17
|
Parekh PJ, Arusi E, Vinik AI, Johnson DA. The role and influence of gut microbiota in pathogenesis and management of obesity and metabolic syndrome. Front Endocrinol (Lausanne) 2014; 5:47. [PMID: 24778627 PMCID: PMC3984999 DOI: 10.3389/fendo.2014.00047] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/23/2014] [Indexed: 12/15/2022] Open
Abstract
The obesity epidemic has drastically impacted the state of health care in the United States. Aside from poor diet hygiene and genetics, there are many other factors thought to play a role in the emergence of obesity and the metabolic syndrome. There has been a paradigm shift toward further investigating the gut microbiota and its implications in the pathogenesis of a variety of disease states, including inflammatory bowel disease, Clostridium difficile, and most recently obesity and the metabolic syndrome. This article is intended to evaluate the role of gut microbiota in the pathogenesis of obesity and metabolic syndrome and its influence in future management.
Collapse
Affiliation(s)
- Parth J. Parekh
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Eli Arusi
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Aaron I. Vinik
- Endocrinology Division, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - David A. Johnson
- Gastroenterology Division, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
- *Correspondence: David A. Johnson, Division of Gastroenterology, Eastern Virginia Medical School, 885 Kempsville Road Suite 114, Norfolk, VA 23502, USA e-mail:
| |
Collapse
|
18
|
Shigwedha N, Sichel L, Jia L, Zhang L. Probiotical Cell Fragments (PCFs) as “Novel Nutraceutical Ingredients”. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/jbm.2014.23007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Sonoda Y, Abdel Mageed AM, Isobe N, Yoshimura Y. Induction of avian β-defensins by CpG oligodeoxynucleotides and proinflammatory cytokines in hen vaginal cells in vitro. Reproduction 2013; 145:621-31. [PMID: 23625580 DOI: 10.1530/rep-12-0518] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Immune function in the vagina of hen oviduct is essential to prevent infection by microorganisms colonizing in the cloaca. The aim of this study was to determine whether CpG oligodeoxynucleotides (CpG-ODN) stimulate the expression of avian β-defensins (AvBDs) in hen vaginal cells. Specific questions were whether CpG-ODN affects the expression of AvBDs and proinflammatory cytokines and whether the cytokines affect AvBDs expression in vaginal cells. The dispersed vaginal cells of White Leghorn laying hens were cultured and stimulated by different doses of lipopolysaccharide (LPS), CpG-ODN, interleukin 1β (IL1B), or IL6. The cultured cell population contained epithelial cells, fibroblast-like cells, and CD45-positive leukocytes. The immunoreactive AvBD3, -10, and -12 were localized in the mucosal epithelium in the section of the vagina. The expression of AvBDs, IL1B, and IL6 was analyzed by quantitative RT-PCR. RT-PCR analysis showed the expression of AvBD1, -3, -4, -5, -10, and -12 in the cultured vaginal cells without stimulation. Toll-like receptors (TLRs) 4 and 21, which recognize LPS and CpG-ODN respectively and IL1 and IL6 receptors (IL1R1 and IL6R) were also expressed in them. The expression of IL1B, IL6, and AvBD10 and -12 was upregulated by LPS, whereas only IL1B and IL6 were upregulated by CpG-ODN. IL1B stimulation upregulated AvBD1 and -3 expression, whereas IL6 stimulation did not cause changes in AvBDs expression. These results suggest that CpG-ODN derived from microbes upregulates the expression of IL1B and IL6 by interaction with TLR21 and then IL1B induces AvBD1 and -3 to prevent infection in the vagina.
Collapse
Affiliation(s)
- Yuka Sonoda
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | | | | | | |
Collapse
|
20
|
Abstract
Current evidence highlights the importance of developing a healthy intestinal microbiota in the neonate. Many aspects that promote health or disease are related to the homeostasis of these intestinal microbiota. Their delicate equilibrium could be strongly influenced by the intervention that physicians perform as part of the medical care of the neonate, especially preterm infants. As awareness of the importance of the development and maintenance of these intestinal flora increase and newer molecular techniques are developed, it will be possible to provide better care of infants with interventions that will have long-lasting effects.
Collapse
|
21
|
|
22
|
Discussion from the 24th Marabou Symposium: Nutrition and the human microbiome. Nutr Rev 2012; 70 Suppl 1:S57-86. [DOI: 10.1111/j.1753-4887.2012.00501.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
23
|
D’Souza A, Cai CL, Kumar D, Cai F, Fordjour L, Ahmad A, Valencia G, Aranda JV, Beharry KD. Cytokines and Toll-like receptor signaling pathways in the terminal ileum of hypoxic/hyperoxic neonatal rats: benefits of probiotics supplementation. Am J Transl Res 2012; 4:187-197. [PMID: 22611471 PMCID: PMC3353534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 03/13/2012] [Indexed: 06/01/2023]
Abstract
BACKGROUND Oxidative stress and inflammation are associated with the development of inflammatory bowel diseases such as necrotizing enterocolitis. We tested the hypothesis that probiotics, prebiotics or synbiotics (a combination of pre- and probiotics) is effective for prevention of inflammatory responses to formula-feeding in the terminal ileum of neonatal rats. METHODS Neonatal rats were exposed to hyperoxia/hypoxia during which they were either maternally-fed or hand-fed with formula supplemented with probiotics, prebiotics or synbiotics. A non-supplemented formula group served as controls. Cytokines and genes associated with oxidative stress and toll-like receptor signaling were determined in the terminal ileum. Data were compared to room air littermates. RESULTS Exposure to hyperoxia/ hypoxia reduced growth accretion in maternally-fed pups. NEC-like symptoms and intestinal inflammatory markers were induced with formula feeding. Supplementation with probiotics, prebiotics or synbiotics decreased proinflammatory cytokines and downregulated genes involved in oxidative stress and toll-like receptor pathways, however, the effect was attenuated in hyperoxia/hypoxia. CONCLUSIONS The combination of suboptimal nutrition due to removal of breast milk and formula feeding with hyperoxia/hypoxia may increase susceptibility to oxidative stress, inflammation, and NEC. Probiotics, prebiotics and synbiotics are protective against oxidative stress and inflammation, but their efficacy may be reduced when administered during hyperoxia/hypoxia insults.
Collapse
Affiliation(s)
- Antoni D’Souza
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Dharmendra Kumar
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Fayme Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Lawrence Fordjour
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
- DeChange of affiliation: Lawrence Fordjour, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Beth Israel Medical CenterManhattan, NY, USA
| | - Asma Ahmad
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Gloria Valencia
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical CenterBrooklyn, NY
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of CaliforniaIrvine, Irvine, CA
| |
Collapse
|
24
|
The role of phosphoinositide 3-kinase signaling in intestinal inflammation. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:358476. [PMID: 22570785 PMCID: PMC3337621 DOI: 10.1155/2012/358476] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 11/29/2011] [Indexed: 02/07/2023]
Abstract
The phosphatidylinositol 3-kinase signaling pathway plays a central role in regulating the host inflammatory response. The net effect can either be pro- or anti-inflammatory depending on the system and cellular context studied. This paper focuses on phosphatidylinositol 3-kinase signaling in innate and adaptive immune cells of the intestinal mucosa. The role of phosphatidylinositol 3-kinase signaling in mouse models of inflammatory bowel disease is also discussed. With the development of new isoform specific inhibitors, we are beginning to understand the specific role of this complex pathway, in particular the role of the γ isoform in intestinal inflammation. Continued research on this complex pathway will enhance our understanding of its role and provide rationale for the design of new approaches to intervention in chronic inflammatory conditions such as inflammatory bowel disease.
Collapse
|
25
|
Wang QY, Sun AM, Song J, Chen Y, Wang JD, Li CG. Cytokine tumor necrosis factor alpha induces intestinal epithelial barrier dysfunction. Cytokine 2012; 58:226-30. [PMID: 22341723 DOI: 10.1016/j.cyto.2012.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 12/30/2011] [Accepted: 01/23/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Epithelial barrier dysfunction is involved in a number of diseases in the body. The mechanism is to be further understood. The present study aimed to investigate the role of one of the common microbial products, flagellin (FGN), in the induction of intestinal epithelial barrier dysfunction. METHODS We collected the colon epithelium specimens from 40 patients with ulcerative colitis (UC), 40 patients with Crohn's disease (CD) and 40 healthy volunteers. The expression of toll like receptors (TLR)5 of the specimens was assessed by RT-PCR and western blotting. The expression of tumor necrosis factor alpha (TNFα) and its role in compromising the barrier function in the intestinal epithelial cells, T84 cells, were observed by a cell culture model. RESULTS The results showed that the expression of TLR5 was observed in the colon epithelium of healthy subjects that was increased in UC patients and further increased in CD patients. Treating T84 cells with FGN increased the expression of TNFα in the cells that caused the T84 cell apoptosis as well as compromised the T84 monolayer barrier function, which could be prevented by knocking down the gene of TNFα in T84 cells. CONCLUSIONS We conclude that the human colon epithelial cells express detectable TLR5 that is increased in patients with CD and UC. The exposure to FGN can increase the expression of TNFα that further compromises the intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Qun-Ying Wang
- Department of Gastroenterology, 401 Hospital of PLA Jinan Military Region, Qingdao, Shandong, China
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW To review interactions between the microbiota and the host in irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), emphasizing areas of commonality and divergence. RECENT FINDINGS Several lines of evidence support a role for the microbiota in the pathogenesis of IBS and IBD. Some implicate the microbiota in a general sense and relate to variations in the composition of the microbiota between IBS, IBD and controls; others relate to the ability of events and interventions that disrupt/modify the microbiota to predispose to the development of IBS and IBD and, others still refer to reports of the ability of antibiotics, prebiotics or probiotics, in selected circumstances, to beneficially alter their clinical course. Enthusiasm for a role for a specific organism in precipitating disease has been largely (and contentiously) linked to IBD. Many issues remain unresolved and must wait for the application of modern microbiological techniques to well characterized populations and well matched controls. SUMMARY It makes sense, given the size and complexity of the microbiota and its role in homeostasis, that the microbiota and its interactions with the host would play a role in the pathogenesis of IBS and IBD; sorting out the details has proven challenging but does offer new therapeutic avenues for both disorders.
Collapse
|