1
|
Chen Y, Wu N, Yan X, Kang L, Ou G, Zhou Z, Xu C, Feng J, Shi T. Impact of gut microbiota on colorectal anastomotic healing (Review). Mol Clin Oncol 2025; 22:52. [PMID: 40297498 PMCID: PMC12035527 DOI: 10.3892/mco.2025.2847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Intestinal anastomosis is a critical procedure in both emergency and elective surgeries to maintain intestinal continuity. However, the incidence of anastomotic leakage (AL) has recently increased, reaching up to 20%, imposing major clinical and economic burdens. Substantial perioperative alterations in the intestinal microbiota composition may contribute to AL, particularly due to disruptions in key microbial populations essential for intestinal health and healing. The intricate interplay between the intestinal microbiota and the host immune system, along with microbial changes before and during surgery, significantly influences anastomotic integrity. Notably, specific pathogens such as Enterococcus and Pseudomonas aeruginosa have been implicated in AL pathogenesis. Preventive strategies including dietary regulation, personalized intestinal preparation, microbiota restoration and enhanced recovery after surgery protocols, may mitigate AL risks. Future research should focus on elucidating the precise mechanisms linking intestinal microbiota alterations to anastomotic healing and developing targeted interventions to improve surgical outcomes.
Collapse
Affiliation(s)
- Yangyang Chen
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Nian Wu
- Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xin Yan
- Anesthesia Operating Room, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Liping Kang
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Guoyong Ou
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Zhenlin Zhou
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Changbo Xu
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Jiayi Feng
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| | - Tou Shi
- General Surgery Department, Guiyang Public Health Clinical Center, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
2
|
Bakke DS, Zhang J, Zhang Y, Ogbu D, Xia Y, Sun J. Myeloid vitamin D receptor regulates Paneth cells and microbial homeostasis. FASEB J 2023; 37:e22957. [PMID: 37219463 PMCID: PMC10321143 DOI: 10.1096/fj.202202169rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023]
Abstract
Cross talk between immune cells and the intestinal crypt is critical in maintaining intestinal homeostasis. Recent studies highlight the direct impact of vitamin D receptor (VDR) signaling on intestinal and microbial homeostasis. However, the tissue-specific role of immune VDR signaling is not fully understood. Here, we generated a myeloid-specific VDR knockout (VDRΔLyz ) mouse model and used a macrophage/enteroids coculture system to examine tissue-specific VDR signaling in intestinal homeostasis. VDRΔLyz mice exhibited small intestine elongation and impaired Paneth cell in maturation and localization. Coculture of enteroids with VDR-/- macrophages increased the delocalization of Paneth cells. VDRΔLyz mice exhibited significant changes in the microbiota taxonomic and functional files, and susceptibility to Salmonella infection. Interestingly, loss of myeloid VDR impaired Wnt secretion in macrophages, thus inhibiting crypt β-catenin signaling and disrupting Paneth cell differentiation in the epithelium. Taken together, our data have demonstrated that myeloid cells regulate crypt differentiation and the microbiota in a VDR-dependent mechanism. Dysregulation of myeloid VDR led to high risks of colitis-associated diseases. Our study provided insight into the mechanism of immune/Paneth cell cross talk in regulating intestinal homeostasis.
Collapse
Affiliation(s)
- Danika S Bakke
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Jilei Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Destiny Ogbu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
- UIC Cancer Center, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
- Jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
3
|
Role of Wnt signaling in the maintenance and regeneration of the intestinal epithelium. Curr Top Dev Biol 2023; 153:281-326. [PMID: 36967198 DOI: 10.1016/bs.ctdb.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The intestinal epithelium plays a key role in digestion and protection against external pathogens. This tissue presents a high cellular turnover with the epithelium being completely renewed every 5days, driven by intestinal stem cells (ISCs) residing in the crypt bases. To sustain this dynamic renewal of the intestinal epithelium, the maintenance, proliferation, and differentiation of ISCs must be precisely controlled. One of the central pathways supporting ISC maintenance and dynamics is the Wnt pathway. In this chapter, we examine the role of Wnt signaling in intestinal epithelial homeostasis and tissue regeneration, including mechanisms regulating ISC identity and fine-tuning of Wnt pathway activation. We extensively discuss the contribution of the stem cell niche in maintaining Wnt signaling in the intestinal crypts that support ISC functions. The integration of these findings highlights the complex interplay of multiple niche signals and cellular components sustaining ISC behavior and maintenance, which together supports the immense plasticity of the intestinal epithelium.
Collapse
|
4
|
Ciaston I, Dobosz E, Potempa J, Koziel J. The subversion of toll-like receptor signaling by bacterial and viral proteases during the development of infectious diseases. Mol Aspects Med 2022; 88:101143. [PMID: 36152458 PMCID: PMC9924004 DOI: 10.1016/j.mam.2022.101143] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/29/2022] [Accepted: 09/09/2022] [Indexed: 02/05/2023]
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors (PRRs) that respond to pathogen-associated molecular patterns (PAMPs). The recognition of specific microbial ligands by TLRs triggers an innate immune response and also promotes adaptive immunity, which is necessary for the efficient elimination of invading pathogens. Successful pathogens have therefore evolved strategies to subvert and/or manipulate TLR signaling. Both the impairment and uncontrolled activation of TLR signaling can harm the host, causing tissue destruction and allowing pathogens to proliferate, thus favoring disease progression. In this context, microbial proteases are key virulence factors that modify components of the TLR signaling pathway. In this review, we discuss the role of bacterial and viral proteases in the manipulation of TLR signaling, highlighting the importance of these enzymes during the development of infectious diseases.
Collapse
Affiliation(s)
- Izabela Ciaston
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Ewelina Dobosz
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jan Potempa
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of Louisville, Louisville, KY, USA.
| | - Joanna Koziel
- Department of Microbiology Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
5
|
Liu Y, Li B, Wei Y. New understanding of gut microbiota and colorectal anastomosis leak: A collaborative review of the current concepts. Front Cell Infect Microbiol 2022; 12:1022603. [PMID: 36389160 PMCID: PMC9663802 DOI: 10.3389/fcimb.2022.1022603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/06/2022] [Indexed: 01/24/2023] Open
Abstract
Anastomotic leak (AL) is a life-threatening postoperative complication following colorectal surgery, which has not decreased over time. Until now, no specific risk factors or surgical technique could be targeted to improve anastomotic healing. In the past decade, gut microbiota dysbiosis has been recognized to contribute to AL, but the exact effects are still vague. In this context, interpretation of the mechanisms underlying how the gut microbiota contributes to AL is significant for improving patients' outcomes. This review concentrates on novel findings to explain how the gut microbiota of patients with AL are altered, how the AL-specific pathogen colonizes and is enriched on the anastomosis site, and how these pathogens conduct their tissue breakdown effects. We build up a framework between the gut microbiota and AL on three levels. Firstly, factors that shape the gut microbiota profiles in patients who developed AL after colorectal surgery include preoperative intervention and surgical factors. Secondly, AL-specific pathogenic or collagenase bacteria adhere to the intestinal mucosa and defend against host clearance, including the interaction between bacterial adhesion and host extracellular matrix (ECM), the biofilm formation, and the weakened host commercial bacterial resistance. Thirdly, we interpret the potential mechanisms of pathogen-induced poor anastomotic healing.
Collapse
Affiliation(s)
- Yang Liu
- Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Science, Ningbo, China,Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, China
| | - Bowen Li
- Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Science, Ningbo, China,Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunwei Wei
- Pancreatic and Gastrointestinal Surgery Division, HwaMei Hospital, University of Chinese Academy of Science, Ningbo, China,Ningbo Clinical Research Center for Digestive System Tumors, Ningbo, China,*Correspondence: Yunwei Wei,
| |
Collapse
|
6
|
Rea V, Bell I, Ball T, Van Raay T. Gut-derived metabolites influence neurodevelopmental gene expression and Wnt signaling events in a germ-free zebrafish model. MICROBIOME 2022; 10:132. [PMID: 35996200 PMCID: PMC9396910 DOI: 10.1186/s40168-022-01302-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Small molecule metabolites produced by the microbiome are known to be neuroactive and are capable of directly impacting the brain and central nervous system, yet there is little data on the contribution of these metabolites to the earliest stages of neural development and neural gene expression. Here, we explore the impact of deriving zebrafish embryos in the absence of microbes on early neural development as well as investigate whether any potential changes can be rescued with treatment of metabolites derived from the zebrafish gut microbiota. RESULTS Overall, we did not observe any gross morphological changes between treatments but did observe a significant decrease in neural gene expression in embryos raised germ-free, which was rescued with the addition of zebrafish metabolites. Specifically, we identified 354 genes significantly downregulated in germ-free embryos compared to conventionally raised embryos via RNA-Seq analysis. Of these, 42 were rescued with a single treatment of zebrafish gut-derived metabolites to germ-free embryos. Gene ontology analysis revealed that these genes are involved in prominent neurodevelopmental pathways including transcriptional regulation and Wnt signaling. Consistent with the ontology analysis, we found alterations in the development of Wnt dependent events which was rescued in the germ-free embryos treated with metabolites. CONCLUSIONS These findings demonstrate that gut-derived metabolites are in part responsible for regulating critical signaling pathways in the brain, especially during neural development. Video abstract.
Collapse
Affiliation(s)
- Victoria Rea
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Ian Bell
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Taylor Ball
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Terence Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada.
| |
Collapse
|
7
|
Abstract
ABSTRACT Accumulating evidence suggests that intestinal bacteria play an important role in the pathogenesis of colorectal cancer (CRC). Due to the complexity of the intestinal microbiome, identification of the specific causative microbial agents in CRC remains challenging, and the search for the causative microbial agents is intense. However, whether bacteria or their products can induce inflammation that results in tumorigenesis or directly causes CRC in humans is still not clear. This review will mainly focus on the progress of bacterial infection and CRC, and introduce the microbial contribution to the hallmarks of cancer. This article uses Salmonella and its chronic infection as an example to investigate a single pathogen and its role in the development of CRC, based on laboratory and epidemiological evidence. The bacterial infection leads to an altered intestinal microbiome. The review also discusses the dysfunction of the microbiome and the mechanism of host-microbial interactions, for example, bacterial virulence factors, key signaling pathways in the host, and microbial post-translational modifications in the tumorigenesis. Colonic carcinogenesis involves a progressive accumulation of mutations in a genetically susceptible host leading to cellular autonomy. Moving forward, more human data are needed to confirm the direct roles of bacterial infection in CRC development. Insights into the inhibiting infection will help to prevent cancer and develop strategies to restore the balance between host and microorganisms.
Collapse
|
8
|
Wnt signaling pathway in cancer immunotherapy. Cancer Lett 2022; 525:84-96. [PMID: 34740608 DOI: 10.1016/j.canlet.2021.10.034] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/06/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022]
Abstract
Wnt/β-catenin signaling is a highly conserved pathway that regulates cell proliferation, differentiation, apoptosis, stem cell self-renewal, tissue homeostasis, and wound healing. Dysregulation of the Wnt pathway is intricately involved in almost all stages of tumorigenesis in various cancers. Through direct and/or indirect effects on effector T cells, T-regulatory cells, T-helper cells, dendritic cells, and other cytokine-expressing immune cells, abnormal activation of Wnt/β-catenin signaling benefits immune exclusion and hinders T-cell-mediated antitumor immune responses. Activation of Wnt signaling results in increased resistance to immunotherapies. In this review, we summarize the process by which Wnt signaling affects cancer and immune surveillance, and the potential for targeting the Wnt-signaling pathway via cancer immunotherapy.
Collapse
|
9
|
Duijster J, Mughini-Gras L, Neefjes J, Franz E. Occupational exposure and risk of colon cancer: a nationwide registry study with emphasis on occupational exposure to zoonotic gastrointestinal pathogens. BMJ Open 2021; 11:e050611. [PMID: 34376453 PMCID: PMC8356182 DOI: 10.1136/bmjopen-2021-050611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES While colon cancer (CC) risk is associated with several lifestyle-related factors, including physical inactivity, smoking and diet, the contribution of occupation to CC morbidity remains largely unclear. Growing evidence indicates that gastrointestinal infections like salmonellosis could contribute to CC development. We performed a nationwide registry study to assess potential associations between occupation (history) and CC, including also those occupations with known increased exposure to gastrointestinal pathogens like Salmonella. METHODS: Person-level occupational data for all residents in The Netherlands were linked to CC diagnosis data. Differences in the incidence of (overall, proximal and distal) CC among occupational sectors and risk groups were tested for significance by calculating standardised incidence ratios (SIRs) with 95% CIs using the general population as reference group. Effects of gender, age, exposure duration and latency were also assessed. RESULTS Significant differences in CC incidence were observed only for a few occupational sectors, including the manufacturing of rubber and plastics, machinery and leather, the printing sector and the information service sector (SIRs 1.06-1.88). No elevated risk of CC was observed among people with increased salmonellosis risk through occupational exposure to live animals, manure or among those working in the sale of animal-derived food products (SIRs 0.93-0.95, 0.81-0.95 and 0.93-1.09 for overall, proximal and distal CC, respectively). CONCLUSIONS The results of this study suggest that occupation in itself provides a relatively small contribution to CC incidence. This is consistent with previous studies where a similar degree of variation in risk estimates was observed. The lack of an association with the high-risk occupations for salmonellosis might be due to higher levels of physical activity, a known protective factor for CC and other diseases, of people working in the agricultural sector, which might outweigh the potential Salmonella-associated risk of CC.
Collapse
Affiliation(s)
- Janneke Duijster
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Cell and Chemical Biology, Oncode Institute, Leiden Universitair Medisch Centrum, Leiden, The Netherlands
| | - Lapo Mughini-Gras
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Institute for Risk Assessment Sciences, Utrecht University Faculty of Veterinary Medicine, Utrecht, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden Universitair Medisch Centrum, Leiden, The Netherlands
| | - Eelco Franz
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
10
|
Botwright NA, Mohamed AR, Slinger J, Lima PC, Wynne JW. Host-Parasite Interaction of Atlantic salmon ( Salmo salar) and the Ectoparasite Neoparamoeba perurans in Amoebic Gill Disease. Front Immunol 2021; 12:672700. [PMID: 34135900 PMCID: PMC8202022 DOI: 10.3389/fimmu.2021.672700] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Marine farmed Atlantic salmon (Salmo salar) are susceptible to recurrent amoebic gill disease (AGD) caused by the ectoparasite Neoparamoeba perurans over the growout production cycle. The parasite elicits a highly localized response within the gill epithelium resulting in multifocal mucoid patches at the site of parasite attachment. This host-parasite response drives a complex immune reaction, which remains poorly understood. To generate a model for host-parasite interaction during pathogenesis of AGD in Atlantic salmon the local (gill) and systemic transcriptomic response in the host, and the parasite during AGD pathogenesis was explored. A dual RNA-seq approach together with differential gene expression and system-wide statistical analyses of gene and transcription factor networks was employed. A multi-tissue transcriptomic data set was generated from the gill (including both lesioned and non-lesioned tissue), head kidney and spleen tissues naïve and AGD-affected Atlantic salmon sourced from an in vivo AGD challenge trial. Differential gene expression of the salmon host indicates local and systemic upregulation of defense and immune responses. Two transcription factors, znfOZF-like and znf70-like, and their associated gene networks significantly altered with disease state. The majority of genes in these networks are candidates for mediators of the immune response, cellular proliferation and invasion. These include Aurora kinase B-like, rho guanine nucleotide exchange factor 25-like and protein NDNF-like inhibited. Analysis of the N. perurans transcriptome during AGD pathology compared to in vitro cultured N. perurans trophozoites, as a proxy for wild type trophozoites, identified multiple gene candidates for virulence and indicates a potential master regulatory gene system analogous to the two-component PhoP/Q system. Candidate genes identified are associated with invasion of host tissue, evasion of host defense mechanisms and formation of the mucoid lesion. We generated a novel model for host-parasite interaction during AGD pathogenesis through integration of host and parasite functional profiles. Collectively, this dual transcriptomic study provides novel molecular insights into the pathology of AGD and provides alternative theories for future research in a step towards improved management of AGD.
Collapse
Affiliation(s)
- Natasha A Botwright
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - Amin R Mohamed
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - Joel Slinger
- Livestock and Aquaculture, CSIRO Agriculture and Food, Woorim, QLD, Australia
| | - Paula C Lima
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - James W Wynne
- Livestock and Aquaculture, CSIRO Agriculture and Food, Hobart, TAS, Australia
| |
Collapse
|
11
|
Gut Microbiota and Colon Cancer: A Role for Bacterial Protein Toxins? Int J Mol Sci 2020; 21:ijms21176201. [PMID: 32867331 PMCID: PMC7504354 DOI: 10.3390/ijms21176201] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence indicates that the human intestinal microbiota can contribute to the etiology of colorectal cancer. Triggering factors, including inflammation and bacterial infections, may favor the shift of the gut microbiota from a mutualistic to a pro-carcinogenic configuration. In this context, certain bacterial pathogens can exert a pro-tumoral activity by producing enzymatically-active protein toxins that either directly induce host cell DNA damage or interfere with essential host cell signaling pathways involved in cell proliferation, apoptosis, and inflammation. This review is focused on those toxins that, by mimicking carcinogens and cancer promoters, could represent a paradigm for bacterially induced carcinogenesis.
Collapse
|
12
|
Jiao Y, Zhang YG, Lin Z, Lu R, Xia Y, Meng C, Pan Z, Xu X, Jiao X, Sun J. Salmonella Enteritidis Effector AvrA Suppresses Autophagy by Reducing Beclin-1 Protein. Front Immunol 2020; 11:686. [PMID: 32362899 PMCID: PMC7181453 DOI: 10.3389/fimmu.2020.00686] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a cellular process to clear pathogens. Salmonella enterica serovar Enteritidis (S.E) has emerged as one of the most important food-borne pathogens. However, major studies still focus on Salmonella enterica serovar Typhimurium. Here, we reported that AvrA, a S. Enteritidis effector, inhibited autophagy to promote bacterial survival in the host. We found that AvrA regulates the conversion of LC3 I into LC3 II and the enrichment of lysosomes. Beclin-1, a key molecular regulator of autophagy, was decreased after AvrA expressed strain colonization. In S.E-AvrA--infected cells, we found the increases of protein levels of p-JNK and p-c-Jun and the transcription level of AP-1. AvrA-reduction of Beclin-1 protein expression is through the JNK pathway. The JNK inhibitor abolished the AvrA-reduced Beclin-1 protein expression. Moreover, we identified that the AvrA mutation C186A abolished its regulation of Beclin-1 expression. In addition AvrA protein was found interacted with Beclin-1. In organoids and infected mice, we explored the physiologically related effects and mechanism of AvrA in reducing Beclin-1 through the JNK pathway, thus attenuating autophagic responses. This finding not only indicates an important role of S. Enteritidis effector in reducing host protein as a strategy to suppress autophagy, but also suggests manipulating autophagy as a new strategy to treat infectious diseases.
Collapse
Affiliation(s)
- Yang Jiao
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Yong-guo Zhang
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Zhijie Lin
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Rong Lu
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Chuang Meng
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Zhimin Pan
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiulong Xu
- Department of Anatomy and Cell Biology, Rush University, Chicago, IL, United States
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Jun Sun
- Division of Gastroenterology and Hepatology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
Infection-induced signals generated at the plasma membrane epigenetically regulate Wnt signaling in vitro and in vivo. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49912-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
14
|
Ahmed I, Roy BC, Rao Jakkula LUM, Subramaniam D, Dandawate P, Anant S, Sampath V, Umar S. Infection-induced signals generated at the plasma membrane epigenetically regulate Wnt signaling in vitro and in vivo. J Biol Chem 2019; 295:1021-1035. [PMID: 31836665 DOI: 10.1074/jbc.ra119.010285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 12/05/2019] [Indexed: 12/14/2022] Open
Abstract
Wnt signaling regulates immunomodulatory functions during infection and inflammation. Employing NCCIT and HCT116 cells, having high endogenous Wnt signaling, we observed elevated levels of low-density lipoprotein receptor-related protein 5/6 (LRP5/6) and Frizzled class receptor 10 (FZD10) and increases in β-catenin, doublecortin-like kinase 1 (DCLK1), CD44 molecule (CD44), and aldehyde dehydrogenase 1 family member A1 (ALDH1A1). siRNA-induced knockdown of these receptors antagonized TOPflash reporter activity and spheroid growth in vitro and elevated Wnt-inhibitory factor 1 (WIF1) activity. Elevated mRNA and protein levels of LRP5/6 and FZD10 paralleled expression of WNT2b and WNT4 in colonic crypts at days 6 and 12 post-infection with Citrobacter rodentium (CR) and tended to decline at days 20-34. The CR mutant escV or the tankyrase inhibitor XAV939 attenuated these responses. A three-dimensional organoid assay in colonic crypts isolated from CR-infected mice revealed elevated levels of LRP5/6 and FZD10 and β-catenin co-localization with enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2). Co-immunoprecipitation in the membrane fraction revealed that axin associates with LRP5/6 in CR-infected crypts, and this association was correlated with increased β-catenin. Colon tumors from either CR-infected ApcP Min/+ or azoxymethane/dextran sodium sulfate (AOM/DSS)-treated mice had high LRP5/6 or FZD10 levels, and chronic Notch blockade through the γ-secretase inhibitor dibenzazepine down-regulated LRP5/6 and FZD10 expression. In CR-responsive CT-26 cells, siRNA-induced LRP5/6 or FZD10 knockdown antagonized TOPflash reporter activity. Elevated miR-153-3p levels correlated with LRP5/6 and FZD10, and miR-153-3p sequestration via a plasmid-based miR inhibitor system attenuated Wnt signaling. We conclude that infection-induced signals from the plasma membrane epigenetically regulate Wnt signaling.
Collapse
Affiliation(s)
- Ishfaq Ahmed
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Badal Chandra Roy
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas 66160
| | | | | | - Prasad Dandawate
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Venkatesh Sampath
- Division of Neonatology, Children's Mercy Hospital, Kansas City, Missouri 64108
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
15
|
Ljungberg JK, Kling JC, Tran TT, Blumenthal A. Functions of the WNT Signaling Network in Shaping Host Responses to Infection. Front Immunol 2019; 10:2521. [PMID: 31781093 PMCID: PMC6857519 DOI: 10.3389/fimmu.2019.02521] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
It is well-established that aberrant WNT expression and signaling is associated with developmental defects, malignant transformation and carcinogenesis. More recently, WNT ligands have emerged as integral components of host responses to infection but their functions in the context of immune responses are incompletely understood. Roles in the modulation of inflammatory cytokine production, host cell intrinsic innate defense mechanisms, as well as the bridging of innate and adaptive immunity have been described. To what degree WNT responses are defined by the nature of the invading pathogen or are specific for subsets of host cells is currently not well-understood. Here we provide an overview of WNT responses during infection with phylogenetically diverse pathogens and highlight functions of WNT ligands in the host defense against infection. Detailed understanding of how the WNT network orchestrates immune cell functions will not only improve our understanding of the fundamental principles underlying complex immune response, but also help identify therapeutic opportunities or potential risks associated with the pharmacological targeting of the WNT network, as currently pursued for novel therapeutics in cancer and bone disorders.
Collapse
Affiliation(s)
- Johanna K Ljungberg
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica C Kling
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Thao Thanh Tran
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Antje Blumenthal
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
16
|
Zhang YG, Zhu X, Lu R, Messer JS, Xia Y, Chang EB, Sun J. Intestinal epithelial HMGB1 inhibits bacterial infection via STAT3 regulation of autophagy. Autophagy 2019; 15:1935-1953. [PMID: 30894054 PMCID: PMC6844505 DOI: 10.1080/15548627.2019.1596485] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 02/20/2019] [Accepted: 03/01/2019] [Indexed: 12/30/2022] Open
Abstract
Extracellular HMGB1 (high mobility group box 1) is considered as a damage-associated molecular pattern protein. However, little is known about its intracellular role. We studied the mechanism whereby intestinal epithelial HMGB1 contributes to host defense, using cell culture, colonoids, conditional intestinal epithelial HMGB1-knockout mice with Salmonella-colitis, il10-/- mice, and human samples. We report that intestinal HMGB1 is an important contributor to host protection from inflammation and infection. We identified a physical interaction between HMGB1 and STAT3. Lacking intestinal epithelial HMGB1 led to redistribution of STAT3 and activation of STAT3 post bacterial infection. Indeed, Salmonella-infected HMGB1-deficient cells exhibited less macroautophagy/autophagy due to decreased expression of autophagy proteins and transcriptional repression by activated STAT3. Then, increased p-STAT3 and extranuclear STAT3 reduced autophagic responses and increased inflammation. STAT3 inhibition restored autophagic responses and reduced bacterial invasion in vitro and in vivo. Moreover, low level of HMGB1 was correlated with reduced nuclear STAT3 and enhanced p-STAT3 in inflamed intestine of il10-/- mice and inflammatory bowel disease (IBD) patients. We revealed that colonic epithelial HMGB1 was directly involved in the suppression of STAT3 activation and the protection of intestine from bacterial infection and injury. Abbreviations: ATG16L1: autophagy-related 16-like 1 (S. cerevisiae); DAMP: damage-associated molecular pattern; HBSS: Hanks balanced salt solution; HMGB1: high mobility group box 1; IBD: inflammatory bowel disease; IL1B/Il-1β: interleukin 1 beta; IL10: interleukin 10; IL17/IL-17: interleukin 17; MEFs: mouse embryonic fibroblasts; STAT3: signal transducer and activator of transcription 3; TLR: toll-like receptor; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Yong-Guo Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Xiaorong Zhu
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Rong Lu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jeannette S. Messer
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Yinglin Xia
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Eugene B. Chang
- Department of Medicine, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
17
|
Intestinal Epithelial Cells Exposed to Bacteroides fragilis Enterotoxin Regulate NF-κB Activation and Inflammatory Responses through β-Catenin Expression. Infect Immun 2019; 87:IAI.00312-19. [PMID: 31451622 DOI: 10.1128/iai.00312-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 08/21/2019] [Indexed: 12/27/2022] Open
Abstract
The Bacteroides fragilis enterotoxin (BFT), a virulence factor of enterotoxigenic B. fragilis (ETBF), interacts with intestinal epithelial cells and can provoke signals that induce mucosal inflammation. Although β-catenin signaling is reported to be associated with inflammatory responses and BFT is known to cleave E-cadherin linked with β-catenin, little is known about the β-catenin-mediated regulation of inflammation in ETBF infection. This study was conducted to investigate the role of β-catenin as a cellular signaling intermediate in the induction of proinflammatory responses to stimulation of intestinal epithelial cells with BFT. Expression of β-catenin in intestinal epithelial cells was reduced relatively early after stimulation with BFT and then recovered to normal levels relatively late after stimulation. In contrast, phosphorylation of β-catenin in BFT-exposed cells occurred at high levels early in stimulation and decreased as time passed. Concurrently, late after stimulation the nuclear levels of β-catenin were relatively higher than those early after stimulation. Suppression of β-catenin resulted in increased NF-κB activity and interleukin-8 (IL-8) expression in BFT-stimulated cells. However, suppression or enhancement of β-catenin expression neither altered the phosphorylated IκB kinase α/β complex nor activated activator protein 1 signals. Furthermore, inhibition of glycogen synthase kinase 3β was associated with increased β-catenin expression and attenuated NF-κB activity and IL-8 expression in BFT-exposed cells. These findings suggest the negative regulation of NF-κB-mediated inflammatory responses by β-catenin in intestinal epithelial cells stimulated with BFT, resulting in attenuation of acute inflammation in ETBF infection.
Collapse
|
18
|
Rogan MR, Patterson LL, Wang JY, McBride JW. Bacterial Manipulation of Wnt Signaling: A Host-Pathogen Tug-of-Wnt. Front Immunol 2019; 10:2390. [PMID: 31681283 PMCID: PMC6811524 DOI: 10.3389/fimmu.2019.02390] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022] Open
Abstract
The host-pathogen interface is a crucial battleground during bacterial infection in which host defenses are met with an array of bacterial counter-mechanisms whereby the invader aims to make the host environment more favorable to survival and dissemination. Interestingly, the eukaryotic Wnt signaling pathway has emerged as a key player in the host and pathogen tug-of-war. Although studied for decades as a regulator of embryogenesis, stem cell maintenance, bone formation, and organogenesis, Wnt signaling has recently been shown to control processes related to bacterial infection in the human host. Wnt signaling pathways contribute to cell cycle control, cytoskeleton reorganization during phagocytosis and cell migration, autophagy, apoptosis, and a number of inflammation-related events. Unsurprisingly, bacterial pathogens have evolved strategies to manipulate these Wnt-associated processes in order to enhance infection and survival within the human host. In this review, we examine the different ways human bacterial pathogens with distinct host cell tropisms and lifestyles exploit Wnt signaling for infection and address the potential of harnessing Wnt-related mechanisms to combat infectious disease.
Collapse
Affiliation(s)
- Madison R. Rogan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - LaNisha L. Patterson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jennifer Y. Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
19
|
Zha L, Garrett S, Sun J. Salmonella Infection in Chronic Inflammation and Gastrointestinal Cancer. Diseases 2019; 7:E28. [PMID: 30857369 PMCID: PMC6473780 DOI: 10.3390/diseases7010028] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/16/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022] Open
Abstract
Salmonella not only causes acute infections, but can also cause patients to become chronic "asymptomatic" carriers. Salmonella has been verified as a pathogenic factor that contributes to chronic inflammation and carcinogenesis. This review summarizes the acute and chronic Salmonella infection and describes the current research progress of Salmonella infection contributing to inflammatory bowel disease and cancer. Furthermore, this review explores the underlying biological mechanism of the host signaling pathways manipulated by Salmonella effector molecules. Using experimental animal models, researchers have shown that Salmonella infection is related to host biological processes, such as host cell transformation, stem cell maintenance, and changes of the gut microbiota (dysbiosis). Finally, this review discusses the current challenges and future directions in studying Salmonella infection and its association with human diseases.
Collapse
Affiliation(s)
- Lang Zha
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Shari Garrett
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
20
|
Abstract
The intestinal epithelial cells function to gain nutrients, retain water and electrolytes, and form an efficient barrier against foreign microbes and antigens. Researchers employed cell culture lines derived from human or animal cancer cells as experimental models in vitro for understanding of intestinal infections. However, most in vitro models used to investigate interactions between bacteria and intestinal epithelial cells fail to recreate the differentiated tissue components and structure observed in the normal intestine. The in vitro analysis of host-bacteria interactions in the intestine has been hampered by a lack of suitable intestinal epithelium culture systems. Here, we present a new experimental model using an organoid culture system to study bacterial infection.
Collapse
Affiliation(s)
- Yong-Guo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, 840 S Wood Street, Chicago, IL, 60612, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, Chicago, IL, 60612, USA.
| |
Collapse
|
21
|
Zhang YG, Lu R, Xia Y, Zhou D, Petrof E, Claud EC, Sun J. Lack of Vitamin D Receptor Leads to Hyperfunction of Claudin-2 in Intestinal Inflammatory Responses. Inflamm Bowel Dis 2019; 25:97-110. [PMID: 30289450 PMCID: PMC6290786 DOI: 10.1093/ibd/izy292] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Vitamin D3 and vitamin D receptor (VDR) are involved in the pathogenesis of inflammatory bowel disease (IBD) and bacterial infection. Claudin-2 is a junction protein that mediates paracellular water transport in epithelia. Elevation of Claudin-2 is associated with active IBD. However, VDR involved in epithelial junctions under inflammation and infection remains largely unknown. We investigated the mechanisms on how VDR and Claudin-2 are related in inflamed states. METHODS Using cultured VDR-/- enteroids, human intestinal epithelial cells, VDR-/- mice with Salmonella- or DSS-colitis, and human IBD samples, we investigated the mechanisms how VDR and Claudin-2 are related in inflamed states. RESULTS After Salmonella infection had taken place, we observed significantly enhanced Claudin-2 and an increased bacterial invasion and translocation. A lack of VDR regulation led to a robust increase of Claudin-2 at the mRNA and protein levels post-infection. In DSS-treated VDR-/- mice, Claudin-2 was significantly increased. Location and quantification of Claudin-2 protein in the mouse colons treated with DSS also confirmed these results. Inflammatory cytokines were significantly higher in the serum and mRNA levels in intestine, which are known to increase Claudin-2. Furthermore, in inflamed intestine of ulcerative colitis patients, VDR expression was low and Claudin-2 was enhanced. Mechanistically, we identified the enhanced Claudin-2 promoter activity through the binding sites of NF-κB and STAT in inflamed VDR-/- cells. CONCLUSIONS Our studies have identified a new role for intestinal epithelial VDR in regulating barrier functions in the context of infection and inflammation.
Collapse
Affiliation(s)
- Yong-guo Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Rong Lu
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yinglin Xia
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - David Zhou
- Department of Pathology, University of Rochester, Rochester, New York, USA,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Elaine Petrof
- Department of Medicine, GI Diseases Research Unit and Division of Infectious Diseases, Queen’s University, Kingston, Ontario, Canada
| | - Erika C Claud
- Department of Pediatrics and Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Address correspondence to: Jun Sun, PhD, AGA Fellow, Professor Division of Gastroenterology and Hepatology Department of Medicine, University of Illinois at Chicago 840 S. Wood Street, Room 704 CSB, MC716 Chicago, IL, 60612, USA. E-mail:
| |
Collapse
|
22
|
Silva PLAPA, Goulart LR, Reis TFM, Mendonça EP, Melo RT, Penha VAS, Peres PABM, Hoepers PG, Beletti ME, Fonseca BB. Biofilm Formation in Different Salmonella Serotypes Isolated from Poultry. Curr Microbiol 2018; 76:124-129. [PMID: 30560366 DOI: 10.1007/s00284-018-1599-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/15/2018] [Indexed: 12/23/2022]
Abstract
Little is known about Salmonella biofilm assembly, making the prevention of the disease a challenge in the poultry production chain. The objective of the present study was then to evaluate biofilm formation from different serotypes of Salmonella spp. in both polystyrene plates and eggshells. Salmonella Gallinarum and S. Minnesota were both classified as producers of biofilms of moderate intensity. Interestingly, S. Gallinarum produces biofilm even though being a serotype without flagellum and not having the lux gene in its genome, suggesting that there might be other important structures and genes associated with biofilm formation. Regarding Enteritidis, Typhimurium, Typhimurium variant, and Heidelberg serotypes, despite having high counts, BFI (Biofilm Formation Index) showed low biofilm production, probably due to the scarcity of extracellular matrix produced by such strains. A turkey eggshell model was then used for S. Enteritidis and S. Heidelberg biofilm formation. The results from the microbial count and scanning electron microscopy showed that Salmonella serotypes were also able to generate biofilm in eggshells, suggesting the presence of biofilms in poultry producing farms, a main concern for the poultry production industry.
Collapse
Affiliation(s)
- Paula L A P A Silva
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Luiz Ricardo Goulart
- Instituto de Genética e Bioquímica da Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Thais F M Reis
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eliane P Mendonça
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Roberta T Melo
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil.
- Faculdade de Medicina Veterinária da Universidade de Uberaba, Uberaba, Brazil.
| | - Victor A S Penha
- Programa de Pós Graduação em Ecologia e Conservação da Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Phelipe A B M Peres
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Patrícia G Hoepers
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
- Animal Health Corporative Area, Foods S/A, Curitiba, PR, Brazil
| | - Marcelo E Beletti
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Belchiolina B Fonseca
- Faculdade de Medicina Veterinária da Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
23
|
van Elsland D, Neefjes J. Bacterial infections and cancer. EMBO Rep 2018; 19:embr.201846632. [PMID: 30348892 PMCID: PMC6216254 DOI: 10.15252/embr.201846632] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/10/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022] Open
Abstract
Infections are estimated to contribute to 20% of all human tumours. These are mainly caused by viruses, which explains why a direct bacterial contribution to cancer formation has been largely ignored. While epidemiological data link bacterial infections to particular cancers, tumour formation is generally assumed to be solely caused by the ensuing inflammation responses. Yet, many bacteria directly manipulate their host cell in various phases of their infection cycle. Such manipulations can affect host cell integrity and can contribute to cancer formation. We here describe how bacterial surface moieties, bacterial protein toxins and bacterial effector proteins can induce host cell DNA damage, and thereby can interfere with essential host cell signalling pathways involved in cell proliferation, apoptosis, differentiation and immune signalling.
Collapse
Affiliation(s)
- Daphne van Elsland
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Leiden, The Netherlands
| | - Jacques Neefjes
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Leiden, The Netherlands
| |
Collapse
|
24
|
Hua K, Li Y, Zhou H, Hu X, Chen Y, He R, Luo R, Zhou R, Bi D, Jin H. Haemophilus parasuis Infection Disrupts Adherens Junctions and Initializes EMT Dependent on Canonical Wnt/β-Catenin Signaling Pathway. Front Cell Infect Microbiol 2018; 8:324. [PMID: 30258822 PMCID: PMC6143654 DOI: 10.3389/fcimb.2018.00324] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
In this study, animal experimentation verified that the canonical Wnt/β-catenin signaling pathway was activated under a reduced activity of p-β-catenin (Ser33/37/Thr41) and an increased accumulation of β-catenin in the lungs and kidneys of pigs infected with a highly virulent strain of H. parasuis. In PK-15 and NPTr cells, it was also confirmed that infection with a high-virulence strain of H. parasuis induced cytoplasmic accumulation and nuclear translocation of β-catenin. H. parasuis infection caused a sharp degradation of E-cadherin and an increase of the epithelial cell monolayer permeability, as well as a broken interaction between β-catenin and E-cadherin dependent on Wnt/β-catenin signaling pathway. Moreover, Wnt/β-catenin signaling pathway also contributed to the initiation of epithelial-mesenchymal transition (EMT) during high-virulence strain of H. parasuis infection with expression changes of epithelial/mesenchymal markers, increased migratory capabilities as well as the morphologically spindle-like switch in PK-15 and NPTr cells. Therefore, we originally speculated that H. parasuis infection activates the canonical Wnt/β-catenin signaling pathway leading to a disruption of the epithelial barrier, altering cell structure and increasing cell migration, which results in severe acute systemic infection characterized by fibrinous polyserositis during H. parasuis infection.
Collapse
Affiliation(s)
- Kexin Hua
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yangjie Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hufeng Zhou
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States.,Department of Immunology and Microbiology, Harvard Medical School, Boston, MA, United States
| | - Xueying Hu
- College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yushan Chen
- Brain and Cognition Research Institute, Wuhan University of Science and Technology, Wuhan, China.,Key Laboratory of Occupational Hazard Identification and Control in Hubei Province, Wuhan University of Science and Technology, Wuhan, China
| | - Rongrong He
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Animal Medicine, Huazhong Agricultural University, Wuhan, China.,Hubei Provincial Key Laboratory of Preventive Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
25
|
Yakymenko O, Schoultz I, Gullberg E, Ström M, Almer S, Wallon C, Wang A, Keita ÅV, Campbell BJ, McKay DM, Söderholm JD. Infliximab restores colonic barrier to adherent-invasive E. coli in Crohn's disease via effects on epithelial lipid rafts. Scand J Gastroenterol 2018; 53:677-684. [PMID: 29688802 DOI: 10.1080/00365521.2018.1458146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Infliximab is important in the therapeutic arsenal of Crohn's disease (CD). However, its effect on mucosal barrier function is not fully understood. Adherent-invasive Escherichia coli (AIEC) are important in CD pathophysiology, but the transmucosal uptake routes are partly unknown. We investigated effects of infliximab on uptake of colon-specific AIEC HM427 across CD colonic mucosa. MATERIALS AND METHODS Endoscopic biopsies from non-inflamed colon of seven patients with CD, before and after two infliximab infusions, and eight non-inflammation controls, were mounted in Ussing chambers. Paracellular permeability (51Cr-EDTA) and transmucosal passage of GFP-expressing HM427 were studied. Mechanisms of HM427 transepithelial transport were investigated in Caco-2 monolayers treated with TNF, in the presence of infliximab and/or endocytosis inhibitors. RESULTS Before infliximab treatment, colonic passage of HM427 [CD: 2475 CFU (450-3000); controls 1163(225-1950)] and 51Cr-EDTA permeability were increased in CD (p < .05), but were restored to control levels by infliximab (CD: 150 (18.8-1069)). In TNF-exposed Caco-2 monolayers HM427 transport and lipid rafts/HM427 co-localization was decreased by infliximab. The lipid raft inhibitor methyl-β-cyclodextrin decreased HM427 transport. CONCLUSION Infliximab restored the colonic barrier to AIEC in CD; an effect partially mediated by blocking lipid rafts in epithelial cells. This ability likely contributes to infliximab's clinical efficacy in colonic CD.
Collapse
Affiliation(s)
- Olena Yakymenko
- a Department of Surgery and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Ida Schoultz
- b Department of Medical Sciences, Faculty of Health and Medicine , Örebro University , Örebro , Sweden
| | - Elisabeth Gullberg
- a Department of Surgery and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Magnus Ström
- c Department of Gastroenterology and Hepatology , Linköping University , Linköping , Sweden
| | - Sven Almer
- d Department of Medicine , Karolinska Institutet , Stockholm , Sweden.,e GastroCentrum , Karolinska University Hospital , Stockholm , Sweden
| | - Conny Wallon
- a Department of Surgery and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Arthur Wang
- f Gastrointestinal Research Group, Cumming School of Medicine , University of Calgary , Calgary , Canada
| | - Åsa V Keita
- a Department of Surgery and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| | - Barry J Campbell
- g Gastroenterology Research Unit, Department of Cellular and Molecular Physiology , University of Liverpool , Liverpool , UK
| | - Derek M McKay
- f Gastrointestinal Research Group, Cumming School of Medicine , University of Calgary , Calgary , Canada
| | - Johan D Söderholm
- a Department of Surgery and Department of Clinical and Experimental Medicine , Linköping University , Linköping , Sweden
| |
Collapse
|
26
|
Novel Regulatory Roles of Wnt1 in Infection-Associated Colorectal Cancer. Neoplasia 2018; 20:499-509. [PMID: 29626750 PMCID: PMC5915993 DOI: 10.1016/j.neo.2018.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 12/11/2022] Open
Abstract
Salmonella infection is a major public health concern, and colonization in humans can be chronic and increases the risk of cancers. Wnt signaling is a key pathway for intestinal renewal and development, inflammation, and tumorigenesis. In the current study, we report a novel role of Wnt1 in infection and colon cancer using cell culture models, a Salmonella-colitis colon cancer model, and human samples. In contrast to the bacteria-induced increases in Wnt2 and Wnt11, Salmonella colonization significantly reduced the level of Wnt1 in intestinal epithelial cells in vivo and in vitro. The bacterial AvrA protein is known to activate the canonical Wnt pathway. Wnt1 expression level was downregulated by AvrA-expressing Salmonella but stabilized by AvrA-deficient Salmonella in the intestine of Salmonella-colitis mice. In a chronic Salmonella-infected cancer model, the Wnt1 protein level was decreased in the AvrA+ infected group. Thus, we further assessed the functional role of Wnt1 downregulation in the inflammatory response and colorectal cancer (CRC) progression. Moreover, downregulation of Wnt1 by the Crispr-Cas9 method promoted cancer cell invasion and migration. Interestingly, we found that Wnt1 was downregulated in human CRC tissue, and Wnt1 downregulation may be correlated with cancer progression. Our study provides insights into mechanisms by which enteric bacteria regulate Wnt1 expression and potentially contribute to infection-associated colon cancer.
Collapse
|
27
|
Microbial interactions with the intestinal epithelium and beyond: Focusing on immune cell maturation and homeostasis. CURRENT PATHOBIOLOGY REPORTS 2018; 6:47-54. [PMID: 30294506 DOI: 10.1007/s40139-018-0165-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Microbial metabolites influence the function of epithelial, endothelial and immune cells in the intestinal mucosa. Microbial metabolites like SCFAs and B complex vitamins direct macrophage polarization whereas microbial derived biogenic amines modulate intestinal epithelium and immune response. Aberrant bacterial lipopolysaccharide-mediated signaling may be involved in the pathogenesis of chronic intestinal inflammation and colorectal carcinogenesis. Our perception of human microbes has changed from that of opportunistic pathogens to active participants maintaining intestinal and whole body homeostasis. This review attempts to explain the dynamic and enriched interactions between the intestinal epithelial mucosa and commensal bacteria in homeostasis maintenance.
Collapse
|
28
|
El Qaidi S, Wu M, Zhu C, Hardwidge PR. Salmonella, E. coli, and Citrobacter Type III Secretion System Effector Proteins that Alter Host Innate Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:205-218. [PMID: 30411307 DOI: 10.1007/5584_2018_289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bacteria deliver virulence proteins termed 'effectors' to counteract host innate immunity. Protein-protein interactions within the host cell ultimately subvert the generation of an inflammatory response to the infecting pathogen. Here we briefly describe a subset of T3SS effectors produced by enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli (EPEC), Citrobacter rodentium, and Salmonella enterica that inhibit innate immune pathways. These effectors are interesting for structural and mechanistic reasons, as well as for their potential utility in being engineered to treat human autoimmune disorders associated with perturbations in NF-κB signaling.
Collapse
Affiliation(s)
- Samir El Qaidi
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Miaomiao Wu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Congrui Zhu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
29
|
Hernández-Nava E, Cuellar P, Nava P, Chávez-Munguía B, Schnoor M, Orozco E, Betanzos A. Adherens junctions and desmosomes are damaged by Entamoeba histolytica: Participation of EhCPADH complex and EhCP112 protease. Cell Microbiol 2017; 19. [PMID: 28656597 DOI: 10.1111/cmi.12761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/08/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022]
Abstract
Entamoeba histolytica trophozoites adhere to epithelium at the cell-cell contact and perturb tight junctions disturbing the transepithelial electrical resistance. Behind tight junctions are the adherens junctions (AJs) that reinforce them and the desmosomes (DSMs) that maintain the epithelium integrity. The damage produced to AJs and DMSs by this parasite is unknown. Here, we studied the effect of the trophozoites, the EhCPADH complex, and the EhCP112 recombinant enzyme (rEhCP112) on AJ and DSM proteins. We found that trophozoites degraded β-cat, E-cad, Dsp l/ll, and Dsg-2 with the participation of EhCPADH and EhCP112. After contact of epithelial cells with trophozoites, immunofluorescence and transmission electron microscopy assays revealed EhCPADH and rEhCP112 at the intercellular space where they colocalised with β-cat, E-cad, Dsp l/ll, and Dsg-2. Moreover, our results suggested that rEhCP112 could be internalised by caveolae and clathrin-coated vesicles. Immunoprecipitation assays showed the interaction of EhCPADH with β-cat and Dsp l/ll. Besides, in vivo assays demonstrated that rEhCP112 concentrates at the cellular borders of the mouse intestine degrading E-cad and Dsp I/II. Our research gives the first clues on the trophozoite attack to AJs and DSMs and point out the role of the EhCPADH and EhCP112 in the multifactorial event of trophozoites virulence.
Collapse
Affiliation(s)
- Elizabeth Hernández-Nava
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Patricia Cuellar
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Porfirio Nava
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Bibiana Chávez-Munguía
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Michael Schnoor
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Abigail Betanzos
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico.,Consejo Nacional de Ciencia y Tecnología, Mexico City, Mexico
| |
Collapse
|
30
|
Xiao Q, Chen Z, Jin X, Mao R, Chen Z. The many postures of noncanonical Wnt signaling in development and diseases. Biomed Pharmacother 2017. [PMID: 28651237 DOI: 10.1016/j.biopha.2017.06.061] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Wnt signaling regulates many aspects of vertebrate development. Its dysregulation causes developmental defects and diseases including cancer. The signaling can be categorized in two pathways: canonical and noncanonical. Canonical pathway plays a key role in regulating proliferation and differentiation of cells whilst noncanonical Wnt signaling mainly controls cellular polarity and motility. During development, noncanonical Wnt signaling is required for tissue formation. Recent studies have shown that noncanonical Wnt signaling is involved in adult tissue development and cancer progression. In this review, we try to describe and discuss the mechanisms behind the biological effects of noncanonical Wnt signaling, diseases caused by its dysregulation, and implications in adult tissue development biology.
Collapse
Affiliation(s)
- Qian Xiao
- Senior Research Scientist, Department of Pharmacology, School of Medicine, Yale University, New Haven, USA
| | - Zhengxi Chen
- PhD, Department of Orthodontics, Ninth People's Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaozhuang Jin
- PhD, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Runyi Mao
- MDS student, Department of Oral and Maxillofacial Surgery, Ninth People's Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenqi Chen
- Professor, Department of Orthodontics, Ninth People's Hospital, School of Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
31
|
Lu R, Wu S, Zhang YG, Xia Y, Zhou Z, Kato I, Dong H, Bissonnette M, Sun J. Salmonella Protein AvrA Activates the STAT3 Signaling Pathway in Colon Cancer. Neoplasia 2017; 18:307-316. [PMID: 27237322 PMCID: PMC4887618 DOI: 10.1016/j.neo.2016.04.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/07/2016] [Indexed: 12/11/2022] Open
Abstract
Salmonella infection in humans can become chronic, which leads to low-grade persistent inflammation. These chronic infections increase the risk of several gastrointestinal diseases, including cancer. Salmonella AvrA is a multifunctional protein that influences eukaryotic cell pathways by regulating ubiquitination and acetylation. In an animal model, we have demonstrated that infection with AvrA-expressing Salmonella induces beta-catenin signals and enhances colonic tumorigenesis. Beta-catenin signaling is a key player in intestinal proliferation and tumorigenesis. The relative contributions of AvrA-induced proliferation and inflammation on tumorigenesis, however, are unknown. STAT3 is activated in chronically inflamed intestines in human inflammatory bowel diseases and in colitis-associated colon cancer. In the current study, mice were colonized with Salmonella AvrA-sufficient or AvrA-deficient bacterial strains. Then, inflammation-associated colon cancer was induced through the use of azoxymethane/dextran sulfate sodium. We determined that AvrA-expressing bacteria activated the STAT3 pathway, which is predicted to enhance proliferation and promote tumorigenesis. Transcriptional activity of STAT3 and its target genes were upregulated by Salmonella expressing AvrA, thus promoting proliferation and intestinal tumorigenesis. Our findings provide new insights regarding a STAT3-dependent mechanism by which the specific bacterial product AvrA enhances the development of infection-associated colon cancer. These insights might suggest future biomarkers to risk assessment and early detection of infection-related cancer.
Collapse
Affiliation(s)
- Rong Lu
- Department of Biochemistry, Rush University, 1735 W Harrison St, Chicago, IL 60612. USA; Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shaoping Wu
- Department of Biochemistry, Rush University, 1735 W Harrison St, Chicago, IL 60612. USA
| | - Yong-Guo Zhang
- Department of Biochemistry, Rush University, 1735 W Harrison St, Chicago, IL 60612. USA; Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yinglin Xia
- Biostatistics and Computational Biology, University of Rochester
| | - Zhongren Zhou
- Pathology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Ikuko Kato
- Department of Oncology and Pathology, Wayne State University School of Medicine, Detroit, MI 48201,USA
| | - Hui Dong
- Department of Gastroenterology, Xingqiao Hospital, The Third Military Medical University, Chongqing 400037, China
| | - Marc Bissonnette
- Department of Medicine, The University of Chicago, Center for Advanced Medicine, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Jun Sun
- Department of Biochemistry, Rush University, 1735 W Harrison St, Chicago, IL 60612. USA; Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
32
|
Gopal A, Chidambaram IS, Devaraj N, Devaraj H. Shigella dysenteriae infection activates proinflammatory response through β-catenin/NF-κB signaling pathway. PLoS One 2017; 12:e0174943. [PMID: 28430783 PMCID: PMC5400225 DOI: 10.1371/journal.pone.0174943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/17/2017] [Indexed: 11/18/2022] Open
Abstract
Shigella dysenteriae (S.dysenteriae) the causative agent of bacillary dysentery invades the human colonic epithelium resulting in severe intestinal inflammatory response and epithelial destruction. However, the mechanism by which S.dysenteriae infection regulates proinflammatory cytokines during intestinal inflammation is still obscure. In this study, we evaluated whether the interaction of β-catenin and NF-κB regulates proinflammatory cytokines TNF-α and IL-8 by modulating GSK-3β activity during S.dysenteriae infection in rat ileal loop model. Here we demonstrated that S.dysenteriae infection stimulate β-catenin degradation which in turn decreased the association between NF-κB and β-catenin. Also, we showed that S.dysenteriae infection increased GSK-3β kinase activity which in turn phosphorylates β-catenin for its degradation by ubiquitination and upregulates IL-8 through NF-κB activation thereby leading to inflammation. Thus these findings revealed the role of β-catenin/ NF-κB and GSK-3β in modulating the inflammatory response during bacterial infection and also showed that β-catenin acts as a critical regulator of inflammation.
Collapse
Affiliation(s)
- Ashidha Gopal
- Unit of Biochemistry, Department of Zoology, University of Madras, Chennai, Tamilnadu, India
| | - Iyer Soumya Chidambaram
- Unit of Biochemistry, Department of Zoology, University of Madras, Chennai, Tamilnadu, India
| | - Niranjali Devaraj
- Department of Biochemistry, University of Madras, Chennai, Tamilnadu, India
| | - Halagowder Devaraj
- Unit of Biochemistry, Department of Zoology, University of Madras, Chennai, Tamilnadu, India
- * E-mail:
| |
Collapse
|
33
|
Bachmann R, Leonard D, Delzenne N, Kartheuser A, Cani PD. Novel insight into the role of microbiota in colorectal surgery. Gut 2017; 66:738-749. [PMID: 28153961 DOI: 10.1136/gutjnl-2016-312569] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/21/2016] [Accepted: 12/24/2016] [Indexed: 12/12/2022]
Abstract
Recent literature undeniably supports the idea that the microbiota has a strong influence on the healing process of an intestinal anastomosis. Understanding the mechanisms by which the bacterial community of the gut influences intestinal healing could open the door for new preventive and therapeutic approaches. Among the different mechanisms, data have shown that the production of specific reactive oxygen species (ROS) and the activation of specific formyl peptide receptors (FPRs) regulate intestinal wound healing. Evidence suggests that specific gut microbes such as Lactobacillus spp and Akkermansia muciniphila help to regulate healing processes through both ROS-dependent and FPR-dependent mechanisms. In this review, we will discuss the current knowledge and future perspectives concerning the impact of microbiota on wound healing. We will further review available evidence on whether mechanical bowel preparation and the use of specific antibiotics are beneficial or harmful procedures, an ongoing matter of debate. These practices have a profound effect on the gut microbiota composition at the level of both the mucosal and the luminal compartments. Therefore, a key question remains unanswered: should we continue to prepare the gut before surgical intervention? Current knowledge and data do not clearly support the use of one technique or another to avoid complications such as anastomotic leak. There is an urgent need for appropriate interventions with a deep microbiota analysis to investigate both the surgical technical benefits of a proper anastomosis compared with the potential effect of the gut microbes (beneficial vs harmful) on the processes of wound healing and anastomotic leakage reduction.
Collapse
Affiliation(s)
- Radu Bachmann
- Colorectal Surgery Unit, Cliniques universitaires Saint-Luc, Brussels, Belgium.,Cliniques des Pathologies Tumorales du Colon et de Rectum (CPTCR), Institut Roi Albert II, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Daniel Leonard
- Colorectal Surgery Unit, Cliniques universitaires Saint-Luc, Brussels, Belgium.,Cliniques des Pathologies Tumorales du Colon et de Rectum (CPTCR), Institut Roi Albert II, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Nathalie Delzenne
- Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition research group, Brussels, Belgium
| | - Alex Kartheuser
- Colorectal Surgery Unit, Cliniques universitaires Saint-Luc, Brussels, Belgium.,Cliniques des Pathologies Tumorales du Colon et de Rectum (CPTCR), Institut Roi Albert II, Cliniques universitaires Saint Luc, Brussels, Belgium
| | - Patrice D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition research group, Brussels, Belgium.,WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Brussels, Belgium
| |
Collapse
|
34
|
YopJ Family Effectors Promote Bacterial Infection through a Unique Acetyltransferase Activity. Microbiol Mol Biol Rev 2016; 80:1011-1027. [PMID: 27784797 DOI: 10.1128/mmbr.00032-16] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gram-negative bacterial pathogens rely on the type III secretion system to inject virulence proteins into host cells. These type III secreted "effector" proteins directly manipulate cellular processes to cause disease. Although the effector repertoires in different bacterial species are highly variable, the Yersinia outer protein J (YopJ) effector family is unique in that its members are produced by diverse animal and plant pathogens as well as a nonpathogenic microsymbiont. All YopJ family effectors share a conserved catalytic triad that is identical to that of the C55 family of cysteine proteases. However, an accumulating body of evidence demonstrates that many YopJ effectors modify their target proteins in hosts by acetylating specific serine, threonine, and/or lysine residues. This unique acetyltransferase activity allows the YopJ family effectors to affect the function and/or stability of their targets, thereby dampening innate immunity. Here, we summarize the current understanding of this prevalent and evolutionarily conserved type III effector family by describing their enzymatic activities and virulence functions in animals and plants. In particular, the molecular mechanisms by which representative YopJ family effectors subvert host immunity through posttranslational modification of their target proteins are discussed.
Collapse
|
35
|
Abstract
Although genes contribute to colorectal cancer, the gut microbiota are an important player. Accumulating evidence suggests that chronic infection and the ensuing inflammation contributes to tumor initiation and tumor progression. A variety of bacterial species and tumor-promoting virulence mechanisms have been investigated. Significant advances have been made in understanding the composition and functional capabilities of the gut microbiota and its roles in cancer. In the current review, we discuss the novel roles of microbiota in the progression of colon cancer. Although microbiota technically include organisms other than bacteria e.g., viruses and fungi, this review will primarily focus on bacteria. We summarize epidemiological studies of human microbiome and colon cancer. We discuss the progress in the scientific understanding of the interplay between the gut microbiota, barrier function, and host responses in experimental models. Further, we discuss the potential application in prevention, diagnosis, and therapy of colon cancer by targeting microbiota. We discuss the challenges lie ahead and the future direction in studying gut microbiome in colon cancer to close the gap between the basic sciences and clinical application.
Collapse
Affiliation(s)
- Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ikuko Kato
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
36
|
Xia Y, Wu S. Tissue inhibitor of metalloproteinase 2 inhibits activation of the β-catenin signaling in melanoma cells. Cell Cycle 2016; 14:1666-74. [PMID: 25839957 DOI: 10.1080/15384101.2015.1030557] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The tissue inhibitor of metalloproteinase (TIMP) family, including TIMP-2, regulates the activity of multifunctional metalloproteinases in pathogenesis of melanoma. The Wnt/β-catenin pathway is constitutively activated and plays a critical role in melanoma progression. However, the relationship between TIMP-2 expression and β-catenin activity is still unclear. We hypothesize that TIMP-2 over expression inhibits the activation of the Wnt/β-catenin pathway in melanoma cells. Protein expression, distribution, and transcriptional activity of β-catenin were assayed in established stable melanoma cell lines: parental A2058 expressing, A2058 T2-1 over-expressing (T2-1), and A2058 T2R-7 under-expressing (T2R-7) TIMP-2. Compared to T2-1 cells at the basal level, T2R-7 showed significantly lower amount protein and weaker immunofluorescence staining of β-catenin. This regulation is through posttranslational level via ubiquitination. Functionally, proliferation and cell growth were lower in T2R-7 compared to A2058 and T2-1. Lithium treatment was used to mimics activation of the Wnt/β-catenin pathway. In T2R-7 cells under-expressing TIMP2, lithium significantly increased total β-catenin, nuclear β-catenin, and its downstream protein phosphor-c-Myc (S62). Nuclear β-catenin staining was enhanced in T2R-7. Beta-catenin transcriptional activity and cell proliferation were also increased significantly. Axins inhibit β-catenin pathway via GSK-3 β. We further found the ratio of p-GSK-3 β (S9) to β-catenin and protein levels of Axins were significantly lower, whereas downstream Wnt 11 was high in T2R-7 treated with lithium. Collectively, the high level of TIMP-2 protein inhibits the activation of the Wnt/β-catenin pathway, thus suppressing proliferation. Insights in the molecular mechanisms of TIMP-2 may provide promising opportunities for anti-proliferative therapeutic intervention.
Collapse
Affiliation(s)
- Yuxuan Xia
- a Department of Biochemistry; Rush University ; Chicago , IL , USA
| | | |
Collapse
|
37
|
Chen K, Fu Q, Li D, Wu Y, Sun S, Zhang X. Wnt3a suppresses Pseudomonas aeruginosa-induced inflammation and promotes bacterial killing in macrophages. Mol Med Rep 2016; 13:2439-46. [PMID: 26846714 PMCID: PMC4768980 DOI: 10.3892/mmr.2016.4869] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 01/11/2016] [Indexed: 12/31/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is a common Gram‑negative bacterium and can cause serious infections, including hospital‑acquired pneumonia, suppurative bacterial keratitis and acute burn wound infection. The pathogenesis of PA infections is closely associated with excessive inflammatory responses and bacterial virulence factors. Wingless‑type MMTV integration site family, member 3A (Wnt3a), an upstream mediator in the canonical Wnt signaling pathway, has been implicated as a regulator of inflammation. However, its role in PA‑induced inflammation and bacterial clearance remains unknown. In the present study, the efficacy of Wnt3a conditioned media (Wnt3a‑CM) was assessed using western blotting and immunofluorescence, which showed that β‑catenin, a downstream molecule of Wnt3a, was upregulated and translocated to the nucleus following exposure to 50% Wnt3a‑CM for 6 h. To explore the role of Wnt3a in PA‑induced inflammation, the mRNA levels of pro‑inflammatory cytokines and apoptosis in macrophages were measured using reverse transcription‑quantitative polymerase chain reaction and flow cytometry, respectively. This indicated that Wnt3a suppressed inflammation by reducing the production of pro‑inflammatory cytokines and by promoting apoptosis in macrophages. Furthermore, the mechanism of macrophage‑mediated bacterial killing was investigated, and the results indicated that Wnt3a enhanced macrophage‑mediated intracellular bacterial killing via the induction of the production of cathelicidin‑related antimicrobial peptide and β‑defensins 1. Taken together, the current study explored the role of Wnt3a in inflammation and bacterial invasion, which may provide an improved understanding of host resistance to PA infection.
Collapse
Affiliation(s)
- Kang Chen
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Qiang Fu
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Dandan Li
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yongjian Wu
- Department of Immunology, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shijun Sun
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| | - Xiumin Zhang
- Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat‑Sen University, Zhongshan, Guangdong 528403, P.R. China
| |
Collapse
|
38
|
Ehrlichia chaffeensis Exploits Canonical and Noncanonical Host Wnt Signaling Pathways To Stimulate Phagocytosis and Promote Intracellular Survival. Infect Immun 2015; 84:686-700. [PMID: 26712203 DOI: 10.1128/iai.01289-15] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/11/2015] [Indexed: 11/20/2022] Open
Abstract
Ehrlichia chaffeensis invades and survives in phagocytes by modulating host cell processes and evading innate defenses, but the mechanisms are not fully defined. Recently we have determined that E. chaffeensis tandem repeat proteins (TRPs) are type 1 secreted effectors involved in functionally diverse interactions with host targets, including components of the evolutionarily conserved Wnt signaling pathways. In this study, we demonstrated that induction of host canonical and noncanonical Wnt pathways by E. chaffeensis TRP effectors stimulates phagocytosis and promotes intracellular survival. After E. chaffeensis infection, canonical and noncanonical Wnt signalings were significantly stimulated during early stages of infection (1 to 3 h) which coincided with dephosphorylation and nuclear translocation of β-catenin, a major canonical Wnt signal transducer, and NFATC1, a noncanonical Wnt transcription factor. In total, the expression of ∼44% of Wnt signaling target genes was altered during infection. Knockdown of TRP120-interacting Wnt pathway components/regulators and other critical components, such as Wnt5a ligand, Frizzled 5 receptor, β-catenin, nuclear factor of activated T cells (NFAT), and major signaling molecules, resulted in significant reductions in the ehrlichial load. Moreover, small-molecule inhibitors specific for components of canonical and noncanonical (Ca(2+) and planar cell polarity [PCP]) Wnt pathways, including IWP-2, which blocks Wnt secretion, significantly decreased ehrlichial infection. TRPs directly activated Wnt signaling, as TRP-coated microspheres triggered phagocytosis which was blocked by Wnt pathway inhibitors, demonstrating a key role of TRP activation of Wnt pathways to induce ehrlichial phagocytosis. These novel findings reveal that E. chaffeensis exploits canonical and noncanonical Wnt pathways through TRP effectors to facilitate host cell entry and promote intracellular survival.
Collapse
|
39
|
|
40
|
Wu S, Yoon S, Zhang YG, Lu R, Xia Y, Wan J, Petrof EO, Claud EC, Chen D, Sun J. Vitamin D receptor pathway is required for probiotic protection in colitis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G341-9. [PMID: 26159695 PMCID: PMC4556945 DOI: 10.1152/ajpgi.00105.2015] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/26/2015] [Indexed: 02/08/2023]
Abstract
Low expression of vitamin D receptor (VDR) and dysfunction of vitamin D/VDR signaling are reported in patients with inflammatory bowel disease (IBD); therefore, restoration of VDR function to control inflammation in IBD is desirable. Probiotics have been used in the treatment of IBD. However, the role of probiotics in the modulation of VDR signaling to effectively reduce inflammation is unknown. We identified a novel role of probiotics in activating VDR activity, thus inhibiting inflammation, using cell models and VDR knockout mice. We found that the probiotics Lactobacillus rhamnosus strain GG (LGG) and Lactobacillus plantarum (LP) increased VDR protein expression in both mouse and human intestinal epithelial cells. Using the VDR luciferase reporter vector, we detected increased transcriptional activity of VDR after probiotic treatment. Probiotics increased the expression of the VDR target genes, such as antimicrobial peptide cathelicidin, at the transcriptional level. Furthermore, the role of probiotics in regulating VDR signaling was tested in vivo using a Salmonella-colitis model in VDR knockout mice. Probiotic treatment conferred physiological and histologic protection from Salmonella-induced colitis in VDR(+/+) mice, whereas probiotics had no effects in the VDR(-/-) mice. Probiotic treatment also enhanced numbers of Paneth cells, which secrete AMPs for host defense. These data indicate that the VDR pathway is required for probiotic protection in colitis. Understanding how probiotics enhance VDR signaling and inhibit inflammation will allow probiotics to be used effectively, resulting in innovative approaches to the prevention and treatment of chronic inflammation.
Collapse
Affiliation(s)
- Shaoping Wu
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Sonia Yoon
- 2Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medical College, Cornell University, New York, New York;
| | - Yong-Guo Zhang
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Rong Lu
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Yinglin Xia
- 3Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York;
| | - Jiandi Wan
- 4Microsystems Engineering, Rochester Institute of Technology, Rochester, New York;
| | - Elaine O. Petrof
- 5Department of Medicine, Gastrointestinal Diseases Research Unit and Division of Infectious Diseases, Queen's University, Kingston, Ontario, Canada; and
| | - Erika C. Claud
- 6Departments of Pediatrics and Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Di Chen
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, Illinois;
| |
Collapse
|
41
|
Zhang Y, Wu S, Ma J, Xia Y, Ai X, Sun J. Bacterial protein AvrA stabilizes intestinal epithelial tight junctions via blockage of the C-Jun N-terminal kinase pathway. Tissue Barriers 2015; 3:e972849. [PMID: 25838979 DOI: 10.4161/21688362.2014.972849] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/27/2014] [Indexed: 12/24/2022] Open
Abstract
The Salmonella type III secretory system secretes virulence proteins, called effectors. Effectors are responsible for the alteration of tight junctions (TJ) and epithelial functions in intestinal infection and inflammation. In a previous study, we have demonstrated that a bacterial effector AvrA plays a role in stabilizing TJs and balancing the opposing action of other bacterial effectors. However, the molecular mechanisms by which AvrA-modulates TJ protein expression remain unknown. AvrA possesses acetyltransferase activity toward specific mitogen-activated protein kinase kinases (MAPKKs) and potently inhibits the c-Jun N-terminal kinase (JNK) pathway in inflammation. Inhibition of the JNK pathway is known to inhibit the TJ protein disassemble. Therefore, we hypothesize that AvrA stabilizes intestinal epithelial TJs via c-Jun and JNK pathway blockage. Using both in vitro and in vivo models, we showed that AvrA targets the c-Jun and JNK pathway that in turn stabilizes TJ protein ZO-1. Inhibition of JNK abolished the effect of AvrA on ZO-1. We further determined that AvrA suppressed the transcription factor activator protein-1, which was regulated by activated JNK. Moreover, we identified the functional domain of AvrA that directly regulated TJs using a series of AvrA mutants. The role of AvrA represents a highly refined bacterial strategy that helps the bacteria survive in the host and dampens the inflammatory response of the host. Our findings have uncovered a novel role of the bacterial protein AvrA in suppressing the inflammatory response of the host through JNK-regulated blockage of epithelial cell barrier function.
Collapse
Key Words
- AP-1, activator protein-1
- AvrA
- CD, crohn's disease
- CFU, colony-forming unit
- DMEM, Dulbecco's modified eagle's medium
- FBS, fetal bovine serum
- HBSS, Hank's balanced salt solutions
- IBD, inflammatory bowel diseases
- JNK
- JNK, c-jun N-terminal kinase
- MAPKKs, mitogen-activated protein kinase kinases
- PBS, phosphate buffered saline
- Salmonella
- TER, transepithelial resistance
- TJ, tight junctions
- TTSS III, type III secretory system
- ZO, zonula occludens
- ZO-1
- c-Jun
- epithelial cell
- inflammation
- intestine
- permeability
- tight junction
Collapse
Affiliation(s)
- Yongguo Zhang
- Rush University; Cohn Research Building; Chicago, IL USA
| | - Shaoping Wu
- Rush University; Cohn Research Building; Chicago, IL USA
| | - Jun Ma
- Rush University; Cohn Research Building; Chicago, IL USA
| | - Yinglin Xia
- Department of Biostatistics and Computational Biology; University of Rochester ; Rochester, NY USA
| | - Xun Ai
- Department of Cell and Molecular Physiology; Stritch School of Medicine ; Loyola University ; Chicago, IL USA
| | - Jun Sun
- Rush University; Cohn Research Building; Chicago, IL USA
| |
Collapse
|
42
|
Alomairi J, Bonacci T, Ghigo E, Soubeyran P. Alterations of host cell ubiquitination machinery by pathogenic bacteria. Front Cell Infect Microbiol 2015; 5:17. [PMID: 25774357 PMCID: PMC4343185 DOI: 10.3389/fcimb.2015.00017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/09/2015] [Indexed: 11/13/2022] Open
Abstract
Response of immune and non-immune cells to pathogens infections is a very dynamic process. It involves the activation/modulation of many pathways leading to actin remodeling, membrane engulfing, phagocytosis, vesicle trafficking, phagolysosome formation, aiming at the destruction of the intruder. These sophisticated and rapid mechanisms rely on post-translational modifications (PTMs) of key host cells' factors, and bacteria have developed various strategies to manipulate them to favor their survival. Among these important PTMs, ubiquitination has emerged as a major mediator/modulator/regulator of host cells response to infections that pathogens have also learned to use for their own benefit. In this mini-review, we summarize our current knowledge about the normal functions of ubiquitination during host cell infection, and we detail its hijacking by model pathogens to escape clearance and to proliferate.
Collapse
Affiliation(s)
- Jaafar Alomairi
- Cellular Stress, Centre de Recherche en Carcérologie de Marseille, INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes Marseille, France ; Infections, Gender and Pregnancy Laboratory, URMITE-IRD198, INSERM U1095, CNRS UMR7278, Aix-Marseille University Marseille, France
| | - Thomas Bonacci
- Cellular Stress, Centre de Recherche en Carcérologie de Marseille, INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes Marseille, France
| | - Eric Ghigo
- Infections, Gender and Pregnancy Laboratory, URMITE-IRD198, INSERM U1095, CNRS UMR7278, Aix-Marseille University Marseille, France
| | - Philippe Soubeyran
- Cellular Stress, Centre de Recherche en Carcérologie de Marseille, INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes Marseille, France
| |
Collapse
|
43
|
Zhang YG, Wu S, Xia Y, Sun J. Salmonella-infected crypt-derived intestinal organoid culture system for host-bacterial interactions. Physiol Rep 2014; 2:2/9/e12147. [PMID: 25214524 PMCID: PMC4270227 DOI: 10.14814/phy2.12147] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The in vitro analysis of bacterial-epithelial interactions in the intestine has been hampered by a lack of suitable intestinal epithelium culture systems. Here, we report a new experimental model using an organoid culture system to study pathophysiology of bacterial-epithelial interactions post Salmonella infection. Using crypt-derived mouse intestinal organoids, we were able to visualize the invasiveness of Salmonella and the morphologic changes of the organoids. Importantly, we reported bacteria-induced disruption of epithelial tight junctions in the infected organoids. In addition, we showed the inflammatory responses through activation of the NF-κB pathway in the organoids. Moreover, our western blot, PCR, and immunofluorescence data demonstrated that stem cell markers (Lgr5 and Bmi1) were significantly decreased by Salmonella infection (determined using GFP-labeled Lgr5 organoids). For the first time, we created a model system that recapitulated a number of observations from in vivo studies of the Salmonella-infected intestine, including bacterial invasion, altered tight junctions, inflammatory responses, and decreased stem cells. We have demonstrated that the Salmonella-infected organoid culture system is a new experimental model suitable for studying host-bacterial interactions.
Collapse
Affiliation(s)
- Yong-Guo Zhang
- Department of Biochemistry, Rush University, 1735 W. Harrison St., Chicago, Illinois
| | - Shaoping Wu
- Department of Biochemistry, Rush University, 1735 W. Harrison St., Chicago, Illinois
| | - Yinglin Xia
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York
| | - Jun Sun
- Department of Biochemistry, Rush University, 1735 W. Harrison St., Chicago, Illinois Department of Internal Medicine (GI), Rush University, Chicago, Illinois Department of Microbiology/Immunology, Rush University, Chicago, Illinois
| |
Collapse
|
44
|
Lu R, Wu S, Zhang YG, Xia Y, Liu X, Zheng Y, Chen H, Schaefer KL, Zhou Z, Bissonnette M, Li L, Sun J. Enteric bacterial protein AvrA promotes colonic tumorigenesis and activates colonic beta-catenin signaling pathway. Oncogenesis 2014; 3:e105. [PMID: 24911876 PMCID: PMC4150214 DOI: 10.1038/oncsis.2014.20] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 02/07/2023] Open
Abstract
Salmonella infections can become chronic and increase the risk of cancer. The mechanisms by which specific Salmonella organisms contribute to cancer, however, are still unknown. Live and attenuated Salmonella are used as vectors to target cancer cells, but there have been no systematic studies of the oncogenic potential of chronic Salmonella infections in cancer models. AvrA, a pathogenic product of Salmonella, is inserted into host cells during infection and influences eukaryotic cell pathways. In the current study, we colonized mice with Salmonella AvrA-sufficient or AvrA-deficient Salmonella typhimirium strains and induced inflammation-associated colon cancer by azoxymethane/dextran sulfate sodium (AOM/DSS). We confirmed Salmonella persisted in the colon for up to 45 weeks. Salmonella was identified not only in epithelial cells on the colonic luminal surface and base of the crypts but also in invading tumors. Tumor incidence in the AvrA+infected group was 100% compared with 51.4% in the AOM/DSS group without bacterial gavage and 56.3% in mice infected with the AvrA- strain. Infection with AvrA+ strain also altered tumor distribution from the distal to proximal colon that might reflect changes in the microbiome. AvrA-expressing bacteria also upregulated beta-catenin signaling as assessed by decreased beta-catenin ubiquitination, increased nuclear beta-catenin and increased phosphorylated-beta-catenin (Ser552), a marker of proliferating stem-progenitor cells. Other β-catenin targets increased by AvrA included Bmi1, a cancer stem cell marker, matrix metalloproteinase-7, and cyclin D1. In summary, AvrA-expressing Salmonella infection activates β-catenin signals and enhances colonic tumorigenesis. Our findings provide important new mechanistic insights into how a bacterial protein targets proliferating stem-progenitor cells and contributes to cancer development. Our observations also raise a note of caution regarding the use of mutant Salmonella organisms as vectors for anti-cancer therapy. Finally, these studies could suggest biomarkers (such as AvrA level in gut) to assess cancer risk in susceptible individuals and infection-related dysregulation of β-catenin signaling in cancer.
Collapse
Affiliation(s)
- R Lu
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - S Wu
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - Y-G Zhang
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - Y Xia
- Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - X Liu
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Y Zheng
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - H Chen
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - K L Schaefer
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Z Zhou
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - M Bissonnette
- Department of Medicine, The University of Chicago, Center for Advanced Medicine, Chicago, IL, USA
| | - L Li
- Stowers Institute, Kansas City, MI, USA
| | - J Sun
- 1] Department of Biochemistry, Rush University, Chicago, IL, USA [2] Department of Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
45
|
Gut commensal bacteria and regional Wnt gene expression in the proximal versus distal colon. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:592-9. [PMID: 24418259 DOI: 10.1016/j.ajpath.2013.11.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/18/2013] [Accepted: 11/22/2013] [Indexed: 02/07/2023]
Abstract
Regional expression of Wingless/Int (Wnt) genes plays a central role in regulating intestinal development and homeostasis. However, our knowledge of such regional Wnt proteins in the colon remains limited. To understand further the effect of Wnt signaling components in controlling intestinal epithelial homeostasis, we investigated whether the physiological heterogeneity of the proximal and distal colon can be explained by differential Wnt signaling. With the use of a Wnt signaling-specific PCR array, expression of 84 Wnt-mediated signal transduction genes was analyzed, and a differential signature of Wnt-related genes in the proximal versus distal murine colon was identified. Several Wnt agonists (Wnt5a, Wnt8b, and Wnt11), the Wnt receptor frizzled family receptor 3, and the Wnt inhibitory factor 1 were differentially expressed along the colon length. These Wnt signatures were associated with differential epithelial cell proliferation and migration in the proximal versus distal colon. Furthermore, reduced Wnt/β-catenin activity and decreased Wnt5a and Wnt11 expression were observed in mice lacking commensal bacteria, an effect that was reversed by conventionalization of germ-free mice. Interestingly, myeloid differentiation primary response gene 88 knockout mice showed decreased Wnt5a levels, indicating a role for Toll-like receptor signaling in regulating Wnt5a expression. Our results suggest that the morphological and physiological heterogeneity within the colon is in part facilitated by the differential expression of Wnt signaling components and influenced by colonization with bacteria.
Collapse
|
46
|
Moossavi S. Location-specific effect of microbiota and MyD88-dependent signaling on Wnt/β-catenin pathway and intestinal stem cells. Gut Microbes 2014; 5:11-4. [PMID: 24424084 PMCID: PMC4049928 DOI: 10.4161/gmic.27291] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Intestinal homeostasis depends on the proper activity of the intestinal stem cells (ISCs) and an appropriate host response to the normal resident microbiota. The question on the effect of microbiota on ISCs behavior has not been addressed yet. Canonical Wnt pathway and ISC gene expression signature was compared in germfree vs. conventional and MyD88(-/-) vs. Myd88(+/+) mice based on publicly available gene expression data sets. Microbiota and MyD88-dependent signaling have distinct effects on the Wnt pathway and ISC at gene expression level. In addition, the effect of microbiota and MyD88-dependent signaling on Wnt pathway and ISCs show regional variation. The net effect of microbiota on Wnt pathway and ISCs cannot be inferred from the available data. Nonetheless, the data are suggestive of a potential regulatory mechanism of the Wnt pathway by the microbiota and plausibly by any alteration in the microbiota composition.
Collapse
|
47
|
Moossavi S, Zhang H, Sun J, Rezaei N. Host-microbiota interaction and intestinal stem cells in chronic inflammation and colorectal cancer. Expert Rev Clin Immunol 2013; 9:409-22. [PMID: 23634736 DOI: 10.1586/eci.13.27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inflammatory bowel disease (IBD) and colorectal cancer (CRC) are the major diseases of the lower gastrointestinal tract. The intestinal epithelium plays a critical role in the host's interactions with the large communities of resident luminal bacteria. Epithelial cells recognize the bacterial components via pattern-recognition receptors. Toll-like receptors (TLRs) are a major class of pattern-recognition receptors that are present on intestinal epithelial cells, including putative stem cells. Stem cells are responsible for tissue homeostasis and regeneration after injury including IBD. Stem cells are also implicated in the pathogenesis of CRC. In susceptible individuals, disruption of normal homeostatic balance between the host's mucosal cells and enteric microflora is believed to result in aberrant immune responses against the resident commensal bacteria, leading to IBD. Microbiological analyses have revealed that the composition and localization of microbiota is altered in CRC and IBD. It is plausible that stem cells directly sense and respond to microbiota. This review aims to summarize the current knowledge on the effect of microbiota and TLR signaling on intestinal stem cells. It also describes how TLR signaling could affect the stem cell regulatory pathways.
Collapse
Affiliation(s)
- Shirin Moossavi
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
48
|
Abstract
Intestinal bacteria can contribute to cell proliferation and cancer development, particularly in chronic infectious diseases in which bacteria and/or bacterial components might interfere with cell function. The number of microbial cells within the gut lumen is estimated to be 100 trillion, which is about 10-times larger than the number of eukaryotic cells in the human body. Because of the complexity of the gut flora, identifying the specific microbial agents related to human diseases remains challenging. Recent studies have demonstrated that the stemness of colon cancer cells is, in part, orchestrated by the microenvironment and is defined by high Wnt activity. In this review article, we will discuss recent progress with respect to intestinal stem cells, cancer stem cells, and the molecular mechanisms of enteric bacteria in the activation of the Wnt pathway. We will also discuss the roles of other pathways, including JAK-STAT, JNK, and Notch, in regulating stem cell niches during bacterial infections using Drosophila models. Insights gained from understanding how host-bacterial interaction during inflammation and cancer may serve as a paradigm for understanding the nature of self-renewal signals.
Collapse
Affiliation(s)
- Jun Sun
- Gastroenterology & Hepatology Division, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA; ; Tel.: +10-585-276-3798
| |
Collapse
|
49
|
Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013; 5:1465-83. [PMID: 24039130 PMCID: PMC3799574 DOI: 10.1002/emmm.201201773] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/19/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022] Open
Abstract
We survive because we adapted to a world of microorganisms. All our epithelial surfaces participate in keeping up an effective barrier against microbes while not initiating ongoing inflammatory processes and risking collateral damage to the host. Major players in this scenario are antimicrobial peptides (AMPs). Such broad-spectrum innate antibiotics are in part produced by specialized cells but also widely sourced from all epithelia as well as circulating inflammatory cells. AMPs belong to an ancient defense system found in all organisms and participated in a preservative co-evolution with a complex microbiome. Particularly interesting interactions between host barrier and microbiota can be found in the gut. The intestinal cell lining not only has to maintain a tightly regulated homeostasis during its high-throughput regeneration, but also a balanced relationship towards an extreme number of mutualistic or commensal inhabitants. Recent research suggests that advancing our understanding of the circumstances of such balanced and sometimes imbalanced interactions between gut microbiota and host AMPs should have therapeutic implications for different intestinal disorders.
Collapse
Affiliation(s)
- Maureen J Ostaff
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Germany
| | | | | |
Collapse
|
50
|
Salmonella infection upregulates the leaky protein claudin-2 in intestinal epithelial cells. PLoS One 2013; 8:e58606. [PMID: 23505542 PMCID: PMC3594366 DOI: 10.1371/journal.pone.0058606] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 02/07/2013] [Indexed: 02/08/2023] Open
Abstract
Background Tight junctions seal the space between adjacent epithelial cells. Mounting evidence suggests that tight junction proteins play a key role in the pathogenesis of human disease. Claudin is a member of the tight junction protein family, which has 24 members in humans. To regulate cellular function, claudins interact structurally and functionally with membrane and scaffolding proteins via their cytoplasmic domain. In particular, claudin-2 is known to be a leaky protein that contributes to inflammatory bowel disease and colon cancer. However, the involvement of claudin-2 in bacterial infection in the intestine remains unknown. Methods/Principal Findings We hypothesized that Salmonella elevates the leaky protein claudin-2 for its own benefit to facilitate bacterial invasion in the colon. Using a Salmonella-colitis mouse model and cultured colonic epithelial cells, we found that pathogenic Salmonella colonization significantly increases the levels of claudin-2 protein and mRNA in the intestine, but not that of claudin-3 or claudin-7 in the colon, in a time-dependent manner. Immunostaining studies showed that the claudin-2 expression along the crypt-villous axis postinfection. In vitro, Salmonella stimulated claudin-2 expression in the human intestinal epithelial cell lines SKCO15 and HT29C19A. Further analysis by siRNA knockdown revealed that claudin-2 is associated with the Salmonella-induced elevation of cell permeability. Epithelial cells with claudin-2 knockdown had significantly less internalized Salmonella than control cells with normal claudin-2 expression. Inhibitor assays demonstrated that this regulation is mediated through activation of the EGFR pathway and the downstream protein JNK. Conclusion/Significance We have shown that Salmonella targets the tight junction protein claudin-2 to facilitate bacterial invasion. We speculate that this disruption of barrier function contributes to a new mechanism by which bacteria interact with their host cells and suggests the possibility of blocking claudin-2 as a potential therapeutic strategy to prevent bacterial invasion.
Collapse
|