1
|
Toris LD, Minsart CF, Husson CP, Franchimont DP, Liefferinckx CL. Lipopolysaccharide-binding protein in Crohn's disease patients: a promising noninvasive biomarker monitoring disease activity. Eur J Gastroenterol Hepatol 2024; 36:1093-1100. [PMID: 38976551 DOI: 10.1097/meg.0000000000002811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
BACKGROUND Following STRIDE-II recommendations, the discovery of novel noninvasive biomarkers, beyond the use of C-reactive protein (CRP) and fecal calprotectin, remains a medical need to further improve the monitoring of patients with inflammatory bowel disease (IBD). This study aims to evaluate the potential of serum lipopolysaccharide-binding protein (LBP) in monitoring IBD activity. METHODS This retrospective cross-sectional study included 69 IBD patients (43 Crohn's disease and 26 ulcerative colitis) and 82 controls. Serum LBP levels were measured by ELISA. Clinical, biological and endoscopic parameters were analyzed for IBD patients with no reports of missing data. Statistical tests, including nonparametric tests and receiver operating characteristic (ROC) curve analysis, were used to evaluate the diagnostic accuracy of LBP. RESULTS IBD patients displayed a significantly higher LBP median [29.6 μg/ml (19.8-38.8) in Crohn's disease and 22.8 (13.7-38.8) in ulcerative colitis] than controls [5.8 (4.7-7.3), P < 0.001] with little overlapping distributions. In Crohn's disease patients, LBP levels gradually increased with endoscopic activity scores demonstrating a 1.7-fold rise in active patients compared to remitter patients ( P = 0.02). LBP level exhibited a positive correlation with CRP ( ρ = 0.75, P < 0.001) as well as fecal calprotectin ( ρ = 0.42, P < 0.01), both of which further increased when excluding cases that did not match endoscopic activity. CONCLUSION LBP might be a promising noninvasive biomarker for monitoring disease activity, especially in Crohn's disease patients. In clinical situations where current biomarkers lack sensitivity, LBP could be discriminative and help filling the gap for reliable therapeutic decisions.
Collapse
Affiliation(s)
- Louison D Toris
- Laboratory of Gastroenterology Experimental (LGE), Université Libre de Bruxelles (ULB)
| | - Charlotte F Minsart
- Laboratory of Gastroenterology Experimental (LGE), Université Libre de Bruxelles (ULB)
- Department of Gastroenterology - Hepato-Pancreatology and Digestive Oncology, Université libre de Bruxelles ULB - Hôpital Universitaire de Bruxelles H.U.B - CUB Hôpital Erasme, Brussels, Belgium
| | - Cécile P Husson
- Laboratory of Gastroenterology Experimental (LGE), Université Libre de Bruxelles (ULB)
| | - Denis P Franchimont
- Laboratory of Gastroenterology Experimental (LGE), Université Libre de Bruxelles (ULB)
- Department of Gastroenterology - Hepato-Pancreatology and Digestive Oncology, Université libre de Bruxelles ULB - Hôpital Universitaire de Bruxelles H.U.B - CUB Hôpital Erasme, Brussels, Belgium
| | - Claire L Liefferinckx
- Laboratory of Gastroenterology Experimental (LGE), Université Libre de Bruxelles (ULB)
- Department of Gastroenterology - Hepato-Pancreatology and Digestive Oncology, Université libre de Bruxelles ULB - Hôpital Universitaire de Bruxelles H.U.B - CUB Hôpital Erasme, Brussels, Belgium
| |
Collapse
|
2
|
Persaud AT, Khela J, Fernandes C, Chaphekar D, Burnie J, Tang VA, Colpitts CC, Guzzo C. Virion-incorporated CD14 enables HIV-1 to bind LPS and initiate TLR4 signaling in immune cells. J Virol 2024; 98:e0036324. [PMID: 38661384 PMCID: PMC11092368 DOI: 10.1128/jvi.00363-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024] Open
Abstract
HIV-1 has a broad range of nuanced interactions with the immune system, and the incorporation of cellular proteins by nascent virions continues to redefine our understanding of the virus-host relationship. Proteins located at the sites of viral egress can be selectively incorporated into the HIV-1 envelope, imparting new functions and phenotypes onto virions, and impacting viral spread and disease. Using virion capture assays and western blot, we show that HIV-1 can incorporate the myeloid antigen CD14 into its viral envelope. Virion-incorporated CD14 remained biologically active and able to bind its natural ligand, bacterial lipopolysaccharide (LPS), as demonstrated by flow virometry and immunoprecipitation assays. Using a Toll-like receptor 4 (TLR4) reporter cell line, we also demonstrated that virions with bound LPS can trigger TLR4 signaling to activate transcription factors that regulate inflammatory gene expression. Complementary assays with THP-1 monocytes demonstrated enhanced secretion of inflammatory cytokines like tumor necrosis factor alpha (TNF-α) and the C-C chemokine ligand 5 (CCL5), when exposed to LPS-loaded virus. These data highlight a new type of interplay between HIV-1 and the myeloid cell compartment, a previously well-established cellular contributor to HIV-1 pathogenesis and inflammation. Persistent gut inflammation is a hallmark of chronic HIV-1 infection, and contributing to this effect is the translocation of microbes across the gut epithelium. Our data herein provide proof of principle that virion-incorporated CD14 could be a novel mechanism through which HIV-1 can drive chronic inflammation, facilitated by HIV-1 particles binding bacterial LPS and initiating inflammatory signaling in TLR4-expressing cells.IMPORTANCEHIV-1 establishes a lifelong infection accompanied by numerous immunological changes. Inflammation of the gut epithelia, exacerbated by the loss of mucosal T cells and cytokine dysregulation, persists during HIV-1 infection. Feeding back into this loop of inflammation is the translocation of intestinal microbes across the gut epithelia, resulting in the systemic dissemination of bacterial antigens, like lipopolysaccharide (LPS). Our group previously demonstrated that the LPS receptor, CD14, can be readily incorporated by HIV-1 particles, supporting previous clinical observations of viruses derived from patient plasma. We now show that CD14 can be incorporated by several primary HIV-1 isolates and that this virion-incorporated CD14 can remain functional, enabling HIV-1 to bind to LPS. This subsequently allowed CD14+ virions to transfer LPS to monocytic cells, eliciting pro-inflammatory signaling and cytokine secretion. We posit here that virion-incorporated CD14 is a potential contributor to the dysregulated immune responses present in the setting of HIV-1 infection.
Collapse
Affiliation(s)
- Arvin T. Persaud
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jasmin Khela
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Claire Fernandes
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Deepa Chaphekar
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Burnie
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vera A. Tang
- Flow Cytometry and Virometry Core Facility, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Che C. Colpitts
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Christina Guzzo
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Pohjonen J, Kaukinen K, Huhtala H, Pörsti I, Lindfors K, Mustonen J, Mäkelä S. Indirect Markers of Intestinal Damage in IgA Nephropathy. Nephron Clin Pract 2024; 148:693-700. [PMID: 38723612 PMCID: PMC11460831 DOI: 10.1159/000538242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/05/2024] [Indexed: 06/11/2024] Open
Abstract
INTRODUCTION Presence of subclinical intestinal inflammation has repeatedly been shown in IgA nephropathy (IgAN) and the degree of histological inflammation has correlated with abnormal urinary findings. There is lack of noninvasive biomarkers evaluating the presence of subclinical intestinal damage in IgAN. We conducted this study hypothesizing that selected biomarkers regarded as indirect markers of intestinal damage could be elevated in IgAN. METHODS Eighty-five primary IgAN patients (median age 55 years, 54% men) participated in this single-center study in Tampere, Finland. None had end-stage kidney disease or previously diagnosed enteropathies. Celiac disease was excluded with serum transglutaminase 2 antibody (TG2Ab) and endomysial antibody tests and inflammatory bowel disease with fecal calprotectin. Intestinal damage was evaluated from sera with analyses of intestinal fatty-acid binding protein (I-FABP), soluble cluster of differentiation molecule 14 (sCD14), and lipopolysaccharide binding protein. Fourteen people suffering from dyspepsia and 15 healthy people served as controls. RESULTS I-FABP levels among IgAN patients were higher than in the healthy controls (median 830 pg/mL vs. 289 pg/mL, p < 0.001). Also, sCD14 was increased in IgAN patients compared to dyspepsia controls. Although TG2Ab levels were within the normal range among IgAN patients, they were higher than in the healthy controls (median 1.3 U/mL vs. 0.6 U/mL, p < 0.001). CONCLUSIONS Elevated serum levels of I-FABP were present in primary IgAN patients without known enteropathies. Serum I-FABP may indicate the presence of subclinical intestinal damage. These findings encourage further investigation into the role of the intestine in the pathophysiology of IgAN.
Collapse
Affiliation(s)
- Jussi Pohjonen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Katri Kaukinen
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Ilkka Pörsti
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jukka Mustonen
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Satu Mäkelä
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
4
|
Mogilevski T, Nguyen AL, Ajamian M, Smith R, Rosella S, Sparrow MP, Moore GT, Gibson PR. Intestinal barrier biomarkers in clinical evaluation of patients with inflammatory bowel disease. Eur J Gastroenterol Hepatol 2024; 36:271-280. [PMID: 38305113 DOI: 10.1097/meg.0000000000002681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is associated with chronic intestinal barrier dysfunction, though its non-invasive assessment remains challenging. This study aimed to determine how four putative circulating markers vary across differing states of intestinal inflammation and with therapy in patients with IBD. METHODS Plasma samples from one prospective cross-sectional and four longitudinal studies, including healthy controls, were analysed for markers of lipopolysaccharide translocation, lipopolysaccharide-binding protein (LBP) and soluble-CD14 (sCD14), and markers of epithelial injury, syndecan-1 and intestinal-type fatty acid-binding protein (IFABP). Inflammatory activity was determined using objective measures. RESULTS Compared with healthy subjects, concentrations of LBP and sCD14 were higher in patients with active (P < 0.001) and severe ulcerative colitis (UC) (P < 0.0001) and active Crohn's disease (CD) (P < 0.001). In UC in remission, LBP was less than in active disease (P = 0.011) LBP levels decreased longitudinally before and after induction of medical therapy in patients with IBD (P = 0.030) and as severe UC was brought into remission at weeks 2 and 12 (P ≤ 0.022). Response to treatment was associated with higher baseline levels of LBP (P = 0.019) and soluble-CD14 (P = 0.014). Concentrations of syndecan-1 and IFABP were or tended to be lower in UC and CD in active disease and did not change with successful therapy. CONCLUSION While markers of epithelial injury were subnormal with active disease and did not change with therapy, markers of lipopolysaccharide translocation directly reflected intestinal inflammation, reduced with successful therapy and predicted treatment response.
Collapse
Affiliation(s)
- Tamara Mogilevski
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
| | - Anke L Nguyen
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
- Gastroenterology Department, Monash Health
| | - Mary Ajamian
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
| | - Rebecca Smith
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
| | - Sam Rosella
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
| | - Miles P Sparrow
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
| | - Gregory T Moore
- Gastroenterology Department, Monash Health
- Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Central Clinical School, Monash University, and Alfred Health
| |
Collapse
|
5
|
Madison AA, Andridge R, Kantaras AH, Renna ME, Bennett JM, Alfano CM, Povoski SP, Agnese DM, Lustberg M, Wesolowski R, Carson WE, Williams NO, Reinbolt RE, Sardesai SD, Noonan AM, Stover DG, Cherian MA, Malarkey WB, Kiecolt-Glaser JK. Depression, Inflammation, and Intestinal Permeability: Associations with Subjective and Objective Cognitive Functioning throughout Breast Cancer Survivorship. Cancers (Basel) 2023; 15:4414. [PMID: 37686689 PMCID: PMC10487080 DOI: 10.3390/cancers15174414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
About one-in-three breast cancer survivors have lingering cognitive complaints and objective cognitive impairment. Chronic inflammation and intestinal permeability (i.e., leaky gut), two risk factors for cognitive decline, can also fuel depression-another vulnerability for cognitive decline. The current study tested whether depression accompanied by high levels of inflammation or intestinal permeability predicted lower subjective and objective cognitive function in breast cancer survivors. We combined data from four breast cancer survivor studies (n = 613); some had repeated measurements for a total of 1015 study visits. All participants had a blood draw to obtain baseline measures of lipopolysaccharide binding protein-a measure of intestinal permeability, as well as three inflammatory markers that were incorporated into an inflammatory index: C-reactive protein, interleukin-6, and tumor necrosis factor-α. They reported depressive symptoms on the Center for Epidemiological Studies depression scale (CES-D), and a binary variable indicated clinically significant depressive symptoms (CES-D ≥ 16). The Kohli (749 observations) and the Breast Cancer Prevention Trial (591 observations) scales assessed subjective cognitive function. Objective cognitive function tests included the trail-making test, Hopkins verbal learning test, Conners continuous performance test, n-back test, FAS test, and animal-naming test (239-246 observations). Adjusting for education, age, BMI, cancer treatment type, time since treatment, study visit, and fatigue, women who had clinically elevated depressive symptoms accompanied by heightened inflammation or intestinal permeability reported poorer focus and marginally poorer memory. However, poorer performance across objective cognitive measures was not specific to inflammation-associated depression. Rather, there was some evidence of lower verbal fluency; poorer attention, verbal learning and memory, and working memory; and difficulties with visuospatial search among depressed survivors, regardless of inflammation. By themselves, inflammation and intestinal permeability less consistently predicted subjective or objective cognitive function. Breast cancer survivors with clinically significant depressive symptoms accompanied by either elevated inflammation or intestinal permeability may perceive greater cognitive difficulty, even though depression-related objective cognitive deficits may not be specific to inflammation- or leaky-gut-associated depression.
Collapse
Affiliation(s)
- Annelise A Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca Andridge
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Biostatistics, The Ohio State University, Columbus, OH 43210, USA
| | - Anthony H Kantaras
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Megan E Renna
- School of Psychology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Jeanette M Bennett
- Department of Psychological Science, University of North Carolina at Charlotte, Charlotte, NC 28213, USA
| | | | - Stephen P Povoski
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Doreen M Agnese
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Maryam Lustberg
- Center for Breast Cancer, Yale Cancer Center, Yale University, New Haven, CT 06519, USA
| | - Robert Wesolowski
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - William E Carson
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nicole O Williams
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Raquel E Reinbolt
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sagar D Sardesai
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anne M Noonan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel G Stover
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mathew A Cherian
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - William B Malarkey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Janice K Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Van den Berghe N, Alsoud D, Verstockt B, Vermeire S, Declerck P, Thomas D. Evaluation of serum cytokines and acute phase proteins as possible pharmacodynamic biomarkers to monitor endoscopic remission during ustekinumab therapy in patients with Crohn's disease. Therap Adv Gastroenterol 2023; 16:17562848231189110. [PMID: 37655059 PMCID: PMC10467236 DOI: 10.1177/17562848231189110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 07/04/2023] [Indexed: 09/02/2023] Open
Abstract
Background Since not all Crohn's disease (CD) patients respond adequately to ustekinumab therapy, biomarkers could aid to monitor treatment response and optimize therapeutic outcomes. Objectives To explore the dynamics of serum biomarker concentrations to monitor the response to ustekinumab treatment in CD patients. Design Retrospective, exploratory study to evaluate concentrations of serum cytokines and acute phase proteins and their relation to endoscopic remission in CD patients during ustekinumab treatment. Methods Serum concentrations of 16 proteins including cytokines and acute phase proteins were measured using the Mesoscale Discovery Platform in serum of healthy controls (n = 13), and CD patients (n = 61) at baseline (week 0), week 8 and week 24 during ustekinumab treatment. Endoscopic remission was defined as simple endoscopic score for CD (SES-CD) <3 after 6 months of therapy. Results Absolute concentrations of serum amyloid A protein (SAA; week 8), IL-6 (week 24), AGP (weeks 8 and 24), interferon (IFN)-γ (weeks 8 and 24), lipopolysaccharide binding protein (LBP; weeks 8 and 24) and IL-22 (weeks 8 and 24) were significantly lower in endoscopic remitters compared to non-responders (p-values ranging between <0.001 and <0.05). SAA (week 8) and AGP (week 24) were the biomarkers with the highest area under the ROC curve (AUROC; 0.761 and 0.760, respectively) for identifying patients in endoscopic remission, though their performance was not superior to C-reactive protein (CRP) or faecal calprotectin. AUROCs of the predictive probability of biomarker combinations showed superiority in discriminating endoscopic remitters from non-responders in comparison to single biomarker measurements, but not as compared to faecal calprotectin. Conclusion Although not superior to faecal calprotectin, measurement of AGP, SAA, LBF, IFN-γ, IL-6 and IL-22 concentrations, and combinations thereof with or without CRP and faecal calprotectin, during ustekinumab therapy might contribute to adequate monitoring of treatment response in CD patients.
Collapse
Affiliation(s)
- Nathalie Van den Berghe
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Dahham Alsoud
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Bram Verstockt
- Tranational Research in Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium – Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium – Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Debby Thomas
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Campus Gasthuisberg O&N2, PB 820, Herestraat 49, Leuven, B-3000, Belgium
| |
Collapse
|
7
|
Zhao Y, Walker DI, Lill CM, Bloem BR, Darweesh SKL, Pinto-Pacheco B, McNeil B, Miller GW, Heath AK, Frissen M, Petrova D, Sánchez MJ, Chirlaque MD, Guevara M, Zibetti M, Panico S, Middleton L, Katzke V, Kaaks R, Riboli E, Masala G, Sieri S, Zamora-Ros R, Amiano P, Jenab M, Peters S, Vermeulen R. Lipopolysaccharide-binding protein and future Parkinson's disease risk: a European prospective cohort. J Neuroinflammation 2023; 20:170. [PMID: 37480114 PMCID: PMC10362572 DOI: 10.1186/s12974-023-02846-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/02/2023] [Indexed: 07/23/2023] Open
Abstract
INTRODUCTION Lipopolysaccharide (LPS) is the outer membrane component of Gram-negative bacteria. LPS-binding protein (LBP) is an acute-phase reactant that mediates immune responses triggered by LPS and has been used as a blood marker for LPS. LBP has recently been indicated to be associated with Parkinson's disease (PD) in small-scale retrospective case-control studies. We aimed to investigate the association between LBP blood levels with PD risk in a nested case-control study within a large European prospective cohort. METHODS A total of 352 incident PD cases (55% males) were identified and one control per case was selected, matched by age at recruitment, sex and study center. LBP levels in plasma collected at recruitment, which was on average 7.8 years before diagnosis of the cases, were analyzed by enzyme linked immunosorbent assay. Odds ratios (ORs) were estimated for one unit increase of the natural log of LBP levels and PD incidence by conditional logistic regression. RESULTS Plasma LBP levels were higher in prospective PD cases compared to controls (median (interquartile range) 26.9 (18.1-41.0) vs. 24.7 (16.6-38.4) µg/ml). The OR for PD incidence per one unit increase of log LBP was elevated (1.46, 95% CI 0.98-2.19). This association was more pronounced among women (OR 2.68, 95% CI 1.40-5.13) and overweight/obese subjects (OR 1.54, 95% CI 1.09-2.18). CONCLUSION The findings suggest that higher plasma LBP levels may be associated with an increased risk of PD and may thus pinpoint to a potential role of endotoxemia in the pathogenesis of PD, particularly in women and overweight/obese individuals.
Collapse
Affiliation(s)
- Yujia Zhao
- Institute for Risk Assessment Sciences, Utrecht University, Nieuw Gildestein, Room 3.53, Yalelaan 2, 3584 CM, Utrecht, The Netherlands
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Christina M Lill
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Bastiaan R Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sirwan K L Darweesh
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brismar Pinto-Pacheco
- Department of Environmental Medicine and Public Health, Icahn School of Medicine, Mount Sinai, New York, USA
| | - Brooklyn McNeil
- Department of Environmental Medicine and Public Health, Icahn School of Medicine, Mount Sinai, New York, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, USA
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Myrthe Frissen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dafina Petrova
- Escuela Andaluza de Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - María-Dolores Chirlaque
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia University, Murcia, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Instituto de Salud Pública y Laboral de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Maurizio Zibetti
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
- SC Neurologia 2U, AOU Città Della Salute E Della Scienza, Turin, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Lefkos Middleton
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
- Public Health Directorate, Imperial College NHS Healthcare Trust, London, UK
| | - Verena Katzke
- Division of Cancer Epidemiology C020, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology C020, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elio Riboli
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, UK
| | - Giovanna Masala
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Pilar Amiano
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Sub Directorate for Public Health and Addictions of Gipuzkoa, Ministry of Health of the Basque Government, San Sebastián, Spain
- Epidemiology of Chronic and Communicable Diseases Group, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Mazda Jenab
- Nutrition and Metabolism (NME) Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Susan Peters
- Institute for Risk Assessment Sciences, Utrecht University, Nieuw Gildestein, Room 3.53, Yalelaan 2, 3584 CM, Utrecht, The Netherlands.
- Institute for Risk Assessment Sciences, Utrecht University, Nieuw Gildestein, Room 3.59, Yalelaan 2, 3584 CM, Utrecht, The Netherlands.
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Nieuw Gildestein, Room 3.53, Yalelaan 2, 3584 CM, Utrecht, The Netherlands.
- University Medical Centre Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
8
|
Gilbert BTP, Lamacchia C, Amend L, Strowig T, Rodriguez E, Palmer G, Finckh A. Brief report: Assessment of mucosal barrier integrity using serological biomarkers in preclinical stages of rheumatoid arthritis. Front Immunol 2023; 14:1117742. [PMID: 36875067 PMCID: PMC9977794 DOI: 10.3389/fimmu.2023.1117742] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Background The pathogenesis of rheumatoid arthritis (RA) is believed to initiate at mucosal sites. The so-called 'mucosal origin hypothesis of RA' postulates an increased intestinal permeability before disease onset. Several biomarkers, including lipopolysaccharide binding protein (LBP) and intestinal fatty acid binding protein (I-FABP), have been proposed to reflect gut mucosa permeability and integrity, while serum calprotectin is a new inflammation marker proposed in RA. Methods We analyzed serum samples of individuals genetically at increased risk of RA in a nested-case-control study. Participants from a longitudinal cohort of first-degree relatives of RA patients (SCREEN-RA cohort) were divided into three pre-clinical stages of RA, based on the presence of risk factors for subsequent RA onset: 1) low-risk healthy asymptomatic controls; 2) intermediate-risk individuals without symptoms, but with RA-associated auto-immunity; 3) high-risk individuals with clinically suspect arthralgias. Five patients with newly diagnosed RA were also sampled. Serum LBP, I-FABP and calprotectin were measured using commercially available ELISA kits. Results We included 180 individuals genetically at increased risk for RA: 84 asymptomatic controls, 53 individuals with RA-associated autoimmunity and 38 high risk individuals. Serum LBP, I-FAPB or calprotectin concentrations did not differ between individuals in different pre-clinical stages of RA. Conclusion Based on the serum biomarkers LBP, I-FABP and calprotectin, we could not detect any evidence for intestinal injury in pre-clinical stages of RA.
Collapse
Affiliation(s)
- Benoît Thomas P Gilbert
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Céline Lamacchia
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lena Amend
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Cluster of Excellence Resolving Infection Susceptibility (RESIST) (EXC 2155), Hannover Medical School, Hannover, Germany.,Center for Individualized Infection Medicine (CiiM), Hannover, Germany
| | - Emiliana Rodriguez
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Axel Finckh
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Inflammation Research (GCIR), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
9
|
Chen SJ, Lin CH. Gut microenvironmental changes as a potential trigger in Parkinson's disease through the gut-brain axis. J Biomed Sci 2022; 29:54. [PMID: 35897024 PMCID: PMC9327249 DOI: 10.1186/s12929-022-00839-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease attributed to the synergistic effects of genetic risk and environmental stimuli. Although PD is characterized by motor dysfunction resulting from intraneuronal alpha-synuclein accumulations, termed Lewy bodies, and dopaminergic neuronal degeneration in the substantia nigra, multiple systems are involved in the disease process, resulting in heterogenous clinical presentation and progression. Genetic predisposition to PD regarding aberrant immune responses, abnormal protein aggregation, autophagolysosomal impairment, and mitochondrial dysfunction leads to vulnerable neurons that are sensitive to environmental triggers and, together, result in neuronal degeneration. Neuropathology studies have shown that, at least in some patients, Lewy bodies start from the enteric nervous system and then spread to the central dopaminergic neurons through the gut-brain axis, suggesting the contribution of an altered gut microenvironment in the pathogenesis of PD. A plethora of evidence has revealed different gut microbiomes and gut metabolites in patients with PD compared to unaffected controls. Chronic gut inflammation and impaired intestinal barrier integrity have been observed in human PD patients and mouse models of PD. These observations led to the hypothesis that an altered gut microenvironment is a potential trigger of the PD process in a genetically susceptible host. In this review, we will discuss the complex interplay between genetic factors and gut microenvironmental changes contributing to PD pathogenesis.
Collapse
Affiliation(s)
- Szu-Ju Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan. .,Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Jackson A, Engen PA, Forsyth CB, Shaikh M, Naqib A, Wilber S, Frausto DM, Raeisi S, Green SJ, Bradaric BD, Persons AL, Voigt RM, Keshavarzian A. Intestinal Barrier Dysfunction in the Absence of Systemic Inflammation Fails to Exacerbate Motor Dysfunction and Brain Pathology in a Mouse Model of Parkinson's Disease. Front Neurol 2022; 13:882628. [PMID: 35665034 PMCID: PMC9159909 DOI: 10.3389/fneur.2022.882628] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/14/2022] [Indexed: 01/01/2023] Open
Abstract
Introduction Parkinson's disease (PD) is the second most common neurodegenerative disease associated with aging. PD patients have systemic and neuroinflammation which is hypothesized to contribute to neurodegeneration. Recent studies highlight the importance of the gut-brain axis in PD pathogenesis and suggest that gut-derived inflammation can trigger and/or promote neuroinflammation and neurodegeneration in PD. However, it is not clear whether microbiota dysbiosis, intestinal barrier dysfunction, or intestinal inflammation (common features in PD patients) are primary drivers of disrupted gut-brain axis in PD that promote neuroinflammation and neurodegeneration. Objective To determine the role of microbiota dysbiosis, intestinal barrier dysfunction, and colonic inflammation in neuroinflammation and neurodegeneration in a genetic rodent model of PD [α-synuclein overexpressing (ASO) mice]. Methods To distinguish the role of intestinal barrier dysfunction separate from inflammation, low dose (1%) dextran sodium sulfate (DSS) was administered in cycles for 52 days to ASO and control mice. The outcomes assessed included intestinal barrier integrity, intestinal inflammation, stool microbiome community, systemic inflammation, motor function, microglial activation, and dopaminergic neurons. Results Low dose DSS treatment caused intestinal barrier dysfunction (sugar test, histological analysis), intestinal microbiota dysbiosis, mild intestinal inflammation (colon shortening, elevated MPO), but it did not increase systemic inflammation (serum cytokines). However, DSS did not exacerbate motor dysfunction, neuroinflammation (microglial activation), or dopaminergic neuron loss in ASO mice. Conclusion Disruption of the intestinal barrier without overt intestinal inflammation is not associated with worsening of PD-like behavior and pathology in ASO mice.
Collapse
Affiliation(s)
- Aeja Jackson
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Phillip A. Engen
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Christopher B. Forsyth
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Maliha Shaikh
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Ankur Naqib
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Sherry Wilber
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Dulce M. Frausto
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Shohreh Raeisi
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Stefan J. Green
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Brinda Desai Bradaric
- Bachelor of Science in Health Sciences Program, College of Health Sciences, Rush University Medical Center, Chicago, IL, United States
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States
| | - Amanda L. Persons
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States
- Department of Physician Assistant Studies, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
11
|
Abstract
Several environmental factors have been implicated in the pathogenesis of inflammatory bowel diseases (IBD); however, the evidence for alcohol is sparse, as is its implications on disease activity and overall management. Here, we examine the available evidence for the effect of alcohol on IBD, including its association with the development of IBD, role in exacerbations, and potential medication interactions. Several mechanisms have been demonstrated to mediate the effects of ethanol in the gastrointestinal tract. Alcohol has been shown to alter the gut microbiome, disrupt intestinal barrier, and increase intestinal permeability, directly and indirectly promoting immune activation. Conversely, specific alcoholic beverages, notably red wine, may have anti-inflammatory properties capable of assisting in disease control and affecting disease monitoring. Nonetheless, most alcohol-mediated effects seem to facilitate intestinal inflammation and consequently impact disease onset, recurrence, and symptom control. Furthermore, alcohol use interferes with the metabolism of several medications leading to increased side effect profiles or even loss of effect. Notably, mesalamine, azathioprine, methotrexate, and biologic medications can all be affected by concomitant alcohol intake via a variety of mechanisms.
Collapse
Affiliation(s)
- Bradley A White
- Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Guilherme Piovezani Ramos
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Sunanda Kane
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
12
|
Oral and Intestinal Bacterial Substances Associated with Disease Activities in Patients with Rheumatoid Arthritis: A Cross-Sectional Clinical Study. J Immunol Res 2022; 2022:6839356. [PMID: 35224112 PMCID: PMC8881124 DOI: 10.1155/2022/6839356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Intestinal bacterial compositions of rheumatoid arthritis (RA) patients have been reported to be different from those of healthy people. Dysbiosis, imbalance of the microbiota, is widely known to cause gut barrier damage, resulting in an influx of bacteria and their substances into host bloodstreams in animal studies. However, few studies have investigated the effect of bacterial substances on the pathophysiology of RA. In this study, eighty-seven active RA patients who had inadequate responses to conventional synthetic disease-modifying antirheumatic drugs or severe comorbidities were analyzed for correlations between many factors such as disease activities, disease biomarkers, intestinal bacterial counts, fecal and serum lipopolysaccharide (LPS), LPS-binding protein (LBP), endotoxin neutralizing capacity (ENC), and serum antibacterial substance IgG and IgA antibody levels by multiple regression analysis with consideration for demographic factors such as age, sex, smoking, and methotrexate treatment. Serum LBP levels, fecal LPS levels, total bacteria counts, serum anti-LPS from Porphyromonas gingivalis (Pg-LPS) IgG antibody levels, and serum anti-Pg-LPS IgA antibody levels were selected for multiple regression analysis using Spearman’s correlation analysis. Serum LBP levels were correlated with disease biomarker levels, such as erythrocyte sedimentation rate (
), C-reactive protein (
), matrix metalloproteinase-3 (
), and IL-6 (
), and were inversely correlated with hemoglobin (
). Anti-Pg-LPS IgG antibody levels were inversely correlated with activity indices such as patient global assessments using visual analogue scale (VAS) (
) and painVAS (
). Total bacteria counts were correlated with ENC (
), and inversely correlated with serum LPS (
) and anti-Pg-LPS IgA antibody levels (
). These results suggest that substances from oral and gut microbiota may influence disease activity in RA patients.
Collapse
|
13
|
Wang R, Deng Y, Zhang Y, Li X, Sun L, Deng Q, Liu Y, Gooneratne R, Li J. Modulation of Intestinal Barrier, Inflammatory Response, and Gut Microbiota by Pediococcus pentosaceus zy-B Alleviates Vibrio parahaemolyticus Infection in C57BL/6J Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1865-1877. [PMID: 35107008 DOI: 10.1021/acs.jafc.1c07450] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Modulation of the intestinal barrier, inflammation, and gut microbiota by Pediococcus pentosaceus zy-B (zy-B) in Vibrio parahaemolyticus (Vp)-infected C57BL/6J mice was studied. Mice intragastrically pretreated with 108 colony-forming units (CFU) zy-B significantly alleviated Vp infection as evidenced by maintaining body weight and reduced disease activity index score and intestine ratio. In addition, zy-B reduced the Vp load in the ileum and cecum, significantly reduced the load in the colon, prevented colonic atrophy, and strengthened mucosal integrity. Mechanistically, zy-B ameliorated intestinal barrier dysfunction by upregulating tight junction protein expression, which in turn reduced the lipopolysaccharide, d-lactic acid (d-LA), and diamine oxidase concentrations and downregulated the cannabinoid receptor 1 (CB1) and CB2 mRNA expressions. Moreover, zy-B systemically reduced inflammation by decreasing interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α levels, and increased interleukin-10 (IL-10), immunoglobulin M (IgM), and immunoglobulin G (IgG) levels in the colon and serum. Furthermore, zy-B markedly altered the gut microbiota composition by enriching Bifidobacterium, Akkermansia, and Lactobacillus in the colon. Overall, zy-B appears to act as a probiotic to alleviate Vp infection by protecting the intestinal barrier, reducing inflammation, and promoting the growth of "beneficial" gut microbiota.
Collapse
Affiliation(s)
- Rundong Wang
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
- College of Food Science and Engineering, Lingnan Normal University, Zhanjiang 524048, China
| | - Yijia Deng
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Xuepeng Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Lijun Sun
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qi Deng
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ying Liu
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury 7647, New Zealand
| | - Jianrong Li
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| |
Collapse
|
14
|
Effect of gluten-free diet on levels of soluble CD14 and lipopolysaccharide-binding protein in adult patients with celiac disease. Cent Eur J Immunol 2021; 46:225-230. [PMID: 34764791 PMCID: PMC8568036 DOI: 10.5114/ceji.2021.107012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/10/2020] [Indexed: 02/05/2023] Open
Abstract
Introduction Celiac disease (CD) is an autoimmune enteropathy triggered by gluten ingestion in genetically susceptible individuals. In CD, activation of the immune response causes damage of the intestinal mucosa, and a gluten-free diet (GFD) is the only available therapy. Intestinal damage can lead to an increase in the circulation of components of bacteria from the intestinal lumen, such as lipopolysaccharide (LPS). Soluble CD14 (sCD14) and lipopolysaccharide-binding protein (LBP) participate in the recognition of LPS, and their levels are altered in different pathologies. In the present study, the circulating levels of sCD14 and LBP from untreated CD patients were evaluated and compared to CD patients on a GFD and controls. Material and methods In total seventy-two adult patients with CD, twenty-three untreated CD patients and forty-nine on a GFD were included. In addition, fifty-five healthy individuals were included as controls. Additionally, the effect of LPS on sCD14 production by both normal and inflamed intestinal tissue culture was explored. Results Serum levels of sCD14 were found to be significantly increased in untreated CD patients compared to patients on a GFD and controls. In addition, we found that LPS induced the production of sCD14 by biopsies of intestinal tissue from untreated CD patients. Conclusions The data from this study show that circulating levels of sCD14 are increased in the untreated CD patients compared to patients on a GFD. Our data show that LPS induces the production of sCD14 by the intestinal tissue from untreated CD patients.
Collapse
|
15
|
Linares R, Francés R, Gutiérrez A, Juanola O. Bacterial Translocation as Inflammatory Driver in Crohn's Disease. Front Cell Dev Biol 2021; 9:703310. [PMID: 34557484 PMCID: PMC8452966 DOI: 10.3389/fcell.2021.703310] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/30/2021] [Indexed: 12/26/2022] Open
Abstract
Crohn’s disease (CD) is a chronic inflammatory disorder of the gastrointestinal tract responsible for intestinal lesions. The multifactorial etiology attributed to CD includes a combination of environmental and host susceptibility factors, which result in an impaired host–microbe gut interaction. Bacterial overgrowth and dysbiosis, increased intestinal barrier permeability, and altered inflammatory responses in patients with CD have been described in the past. Those events explain the pathogenesis of luminal translocation of bacteria or its products into the blood, a frequent event in CD, which, in turn, favors a sustained inflammatory response in these patients. In this review, we navigate through the interaction between bacterial antigen translocation, permeability of the intestinal barrier, immunologic response of the host, and genetic predisposition as a combined effect on the inflammatory response observed in CD. Several lines of evidence support that translocation of bacterial products leads to uncontrolled inflammation in CD patients, and as a matter of fact, the presence of gut bacterial genomic fragments at a systemic level constitutes a marker for increased risk of relapse among CD patients. Also, the significant percentage of CD patients who lose response to biologic therapies may be influenced by the translocation of bacterial products, which are well-known drivers of proinflammatory cytokine production by host immune cells. Further mechanistic studies evaluating cellular and humoral immune responses, gut microbiota alterations, and genetic predisposition will help clinicians to better control and personalize the management of CD patients in the future.
Collapse
Affiliation(s)
- Raquel Linares
- Hepatic and Intestinal Immunobiology Group, Department of Clinical Medicine, Miguel Hernández University, San Juan de Alicante, Spain
| | - Rubén Francés
- Hepatic and Intestinal Immunobiology Group, Department of Clinical Medicine, Miguel Hernández University, San Juan de Alicante, Spain.,CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Instituto ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Ana Gutiérrez
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.,Instituto ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain.,Servicio de Medicina Digestiva, Hospital General Universitario de Alicante, Alicante, Spain
| | - Oriol Juanola
- Translational Research Laboratory, Gastroenterology and Hepatology, Ente Ospedaliero Cantonale, Lugano, Switzerland.,Faculty of Biomedical Sciences, Universitá della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
16
|
Karow F, Smiljanovic B, Grün JR, Poddubnyy D, Proft F, Talpin A, Hue C, Boland A, Deleuze JF, Garchon HJ, Ergenç I, De Craemer AS, Erben U, Häupl T, Elewaut D, Breban M, Grützkau A, Syrbe U. Monocyte transcriptomes from patients with axial spondyloarthritis reveal dysregulated monocytopoiesis and a distinct inflammatory imprint. Arthritis Res Ther 2021; 23:246. [PMID: 34560894 PMCID: PMC8461951 DOI: 10.1186/s13075-021-02623-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In patients with axial spondyloarthritis (axSpA), monocytes show a pre-activated phenotype. Gut inflammation is a trigger of monocyte activation and may also affect their development in the bone marrow (BM). As gut inflammation is commonly observed in axSpA patients, we performed a detailed analysis of monocyte transcriptomes of axSpA patients in two cohorts and searched for signs of activation and developmental adaptations as putative imprints of gut inflammation. METHODS Transcriptomes of blood CD14+ monocytes of HLA-B27+ axSpA patients and healthy controls (HC) were generated by microarrays from cohort 1 and by RNA-sequencing from cohort 2. Differentially expressed genes from both analyses were subjected to gene set enrichment analysis (GSEA) and to co-expression analysis in reference transcriptomes from BM cells, blood cells and activated monocytes. As serological markers of translocation, 1,3 beta-glycan, intestinal fatty acid binding protein, and lipopolysaccharide binding protein (LBP) were determined by LAL and ELISA. RESULTS Transcriptome analysis identified axSpA-specific monocyte signatures showing an imprint of LPS/cytokine-activated monocytes, late granulopoietic BM cells, blood neutrophils, and G-CSF-mobilized blood cells, which suggests LPS/TNF activation and more prominent BM adaptation promoting a neutrophil-like phenotype. GSEA mapped axSpA upregulated genes to inflammatory responses and TNFα signaling and downregulated probe-sets to metabolic pathways. Among translocation markers, LBP levels were significantly increased in axSpA patients vs. HC (p < 0.001). Stratified analysis by disease activity and stage identified an "active disease signature" (BASDAI ≥ 4) with an imprint of LPS/cytokine-activated monocytes and CD16+ monocyte subsets. The "AS signature" (vs. non-radiographic axSpA) showed a reinforced neutrophil-like phenotype due to deprivation of dendritic cell transcripts. CONCLUSIONS The neutrophil-like phenotype of axSpA monocytes points towards a biased monocytopoiesis from granulocyte-monocyte progenitors. This shift in monocytopoiesis and the LPS/cytokine imprint as well as the elevated LBP levels are indicators of systemic inflammation, which may result from bacterial translocation. The BM adaptation is most prominent in AS patients while disease activity appears to be linked to activation and trafficking of monocytes.
Collapse
Affiliation(s)
- Fabian Karow
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Biljana Smiljanovic
- Medizinische Klinik für Rheumatologie und Klinische Immunologie, Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim R Grün
- Deutsches Rheumaforschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Denis Poddubnyy
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Fabian Proft
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Alice Talpin
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d'Excellence Inflamex, Universite de Paris, Paris, France
| | - Christophe Hue
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d'Excellence Inflamex, Universite de Paris, Paris, France
| | - Anne Boland
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- CEA, Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, Evry, France
| | - Jean-François Deleuze
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- CEA, Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, Evry, France
| | - Henri-Jean Garchon
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d'Excellence Inflamex, Universite de Paris, Paris, France
- Service de Biochimie, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Ilkay Ergenç
- VIB Center for Inflammation Research, Ghent University Hospital, Ghent, Belgium
- Department of Rheumatology, Ghent University, Ghent, Belgium
| | - Ann-Sophie De Craemer
- VIB Center for Inflammation Research, Ghent University Hospital, Ghent, Belgium
- Department of Rheumatology, Ghent University, Ghent, Belgium
| | - Ulrike Erben
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Thomas Häupl
- Medizinische Klinik für Rheumatologie und Klinische Immunologie, Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dirk Elewaut
- VIB Center for Inflammation Research, Ghent University Hospital, Ghent, Belgium
- Department of Rheumatology, Ghent University, Ghent, Belgium
| | - Maxime Breban
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d'Excellence Inflamex, Universite de Paris, Paris, France
- Service de Rhumatologie, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Andreas Grützkau
- Deutsches Rheumaforschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Uta Syrbe
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany.
| |
Collapse
|
17
|
Dowdell AS, Colgan SP. Metabolic Host-Microbiota Interactions in Autophagy and the Pathogenesis of Inflammatory Bowel Disease (IBD). Pharmaceuticals (Basel) 2021; 14:708. [PMID: 34451805 PMCID: PMC8399382 DOI: 10.3390/ph14080708] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a family of conditions characterized by chronic, relapsing inflammation of the gastrointestinal tract. IBD afflicts over 3 million adults in the United States and shows increasing prevalence in the Westernized world. Current IBD treatments center on modulation of the damaging inflammatory response and carry risks such as immunosuppression, while the development of more effective treatments is hampered by our poor understanding of the molecular mechanisms of IBD pathogenesis. Previous genome-wide association studies (GWAS) have demonstrated that gene variants linked to the cellular response to microorganisms are most strongly associated with an increased risk of IBD. These studies are supported by mechanistic work demonstrating that IBD-associated polymorphisms compromise the intestine's anti-microbial defense. In this review, we summarize the current knowledge regarding IBD as a disease of defects in host-microbe interactions and discuss potential avenues for targeting this mechanism for future therapeutic development.
Collapse
Affiliation(s)
| | - Sean P. Colgan
- Department of Medicine and the Mucosal Inflammation Program, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| |
Collapse
|
18
|
Chen SJ, Chi YC, Ho CH, Yang WS, Lin CH. Plasma Lipopolysaccharide-Binding Protein Reflects Risk and Progression of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2021; 11:1129-1139. [PMID: 33720853 DOI: 10.3233/jpd-212574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Lipopolysaccharide-binding protein (LBP) presents bacterial endotoxin, lipopolysaccharides, to cellular surface pattern receptors for immune responses in the gut-brain axis of Parkinson's disease (PD). OBJECTIVE We investigated whether plasma LBP levels were associated with PD severity and progression. METHODS This study included 397 participants (248 PD patients and 149 controls). We measured participants' plasma levels of LBP and pro-inflammatory cytokines, including TNF-α, IL-6, andIL-17A. PD patients underwent motor and cognition evaluations at baseline and at a mean follow-up interval of 4.7±2.3 years. We assessed the progression of motor and cognition symptoms based on changes in the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) part III motor score and Mini-Mental State Examination (MMSE) score, respectively. RESULTS Plasma LBP levels were lower in PD patients than controls (9.08±2.91 vs. 10.10±3.00μg/ml, p < 0.01). A multiple logistic regression model with adjustment for age, sex, and plasma cytokine levels revealed that reduced plasma LBP levels were associated with increased PD risk (odds ratio 0.816, [95% CI 0.717-0.929], p = 0.002). Among PD patients, LBP levels were correlated with MDS-UPDRS part III motor score after adjustment for confounders (coefficient = 0.636, p = 0.017), but not with MMSE score. Adjusted Cox regression analysis showed that higher plasma LBP levels associated with faster motor progression (adjusted hazard ratio 1.084 [95% CI 1.011-1.163], p = 0.024) during follow-up. CONCLUSION Our results demonstrated that plasma LBP levels reflect risk, motor symptom severity and progression in patients with PD.
Collapse
Affiliation(s)
- Szu-Ju Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | - Yu-Chiao Chi
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chang-Han Ho
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Shiung Yang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
19
|
Farias e Silva K, Nanini HF, Cascabulho CM, Rosas SLB, Santana PT, Carneiro AJDV, Anaissie E, Nucci M, de Souza HSP. Serum 1,3-beta-D-glucan as a noninvasive test to predict histologic activity in patients with inflammatory bowel disease. World J Gastroenterol 2021; 27:866-885. [PMID: 33727775 PMCID: PMC7941859 DOI: 10.3748/wjg.v27.i9.866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/11/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND 1,3-beta-D-glucan (BG) is a ubiquitous cell wall component of gut micro-organisms. We hypothesized that the serum levels of BG could reflect active intestinal inflammation in patients with inflammatory bowel disease.
AIM To determine whether the serum BG concentrations correlate with intestinal inflammation.
METHODS A prospective observational study was performed in a tertiary referral center, from 2016 to 2019, in which serum BG was determined in 115 patients with Crohn’s disease (CD), 51 with ulcerative colitis (UC), and 82 controls using a photometric detection kit. Inflammatory activity was determined by ileocolonoscopy, histopathology, magnetic resonance enterography, and biomarkers, including fecal calprotectin (FC), C-reactive protein, and a panel of cytokines. The ability of BG to detect active vs inactive disease was assessed using the area under the receiver operating characteristic curve. In subgroup analysis, serial BG was used to assess the response to therapeutic interventions.
RESULTS The serum BG levels were higher in CD patients than in controls (P = 0.0001). The BG levels paralleled the endoscopic activity in CD patients and histologic activity and combined endoscopic and histologic activity in both CD and UC patients. The area under the curve (AUC) in receiver operating characteristic analysis to predict endoscopic activity was 0.694 [95% confidence interval (CI): 0.60-0.79; P = 0.001] in CD, and 0.662 (95%CI: 0.51-0.81; P = 0.066) in UC patients. The AUC in receiver operating characteristic analysis to predict histologic activity was 0.860 (95%CI: 0.77-0.95; P < 0.001) in CD, and 0.786 (95%CI: 0.57-0.99; P = 0.015) in UC patients. The cut-off values of BG for both endoscopic and histologic activity were 60 µg/mL in CD, and 40 µg/mL in UC patients. Performance analysis showed that the results based on BG of 40 and 60 µg/mL were more specific for predicting endoscopic activity (71.8% and 87.2% for CD; and 87.5% and 87.5% for UC, respectively) than FC (53.3% and 66.7% for CD; and 20% and 80% for UC, respectively); and also histologic activity (60.5% and 76.3% for CD; and 90.0% and 95.0% for UC, respectively) than FC (41.7% and 50.0% for CD; and 25% and 50% for UC, respectively). Regarding the clinical, endoscopic, and histologic activities, the BG levels were reduced following therapeutic intervention in patients with CD (P < 0.0001) and UC (P = 0.003). Compared with endoscopic (AUC: 0.693; P = 0.002) and histologic (AUC: 0.868; P < 0.001) activity, no significant correlation was found between serum BG and transmural healing based on magnetic resonance enterography (AUC: 0.576; P = 0.192). Positive correlations were detected between BG and IL-17 in the CD (r: 0.737; P = 0.001) and the UC group (r: 0.574; P = 0.005), and between BG and interferon-gamma in the CD group (r: 0.597; P = 0.015).
CONCLUSION Serum BG may represent an important novel noninvasive approach for detecting mucosal inflammation and therapeutically monitoring inflammatory bowel diseases, particularly in CD.
Collapse
Affiliation(s)
- Katia Farias e Silva
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Hayandra F Nanini
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Cynthia Machado Cascabulho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Instituto Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Siane L B Rosas
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Patricia T Santana
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Antonio José de V Carneiro
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Elias Anaissie
- Clinical Trial and Consulting Services, Cincinnati, OH 45267, United States
| | - Marcio Nucci
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Heitor Siffert Pereira de Souza
- Department of Clinical Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
- Internal Medicine, D'Or Institute for Research and Education (IDOR), Rio de Janeiro 22281-100, Brazil
| |
Collapse
|
20
|
Komarow HD, Brenchley JM, Eisch AR, Young ML, Scott LM, Kulinski JM, Heller T, Bai Y, Metcalfe DD. A study of microbial translocation markers in mastocytosis. Clin Exp Allergy 2021; 51:369-372. [PMID: 33259149 PMCID: PMC8932442 DOI: 10.1111/cea.13798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Hirsh D. Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Robin Eisch
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael L. Young
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Linda M. Scott
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joseph M. Kulinski
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theo Heller
- Translational Hepatology Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dean D. Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
21
|
Ma W, Strate LL, Chan AT. Reply. Clin Gastroenterol Hepatol 2021; 19:208-209. [PMID: 33339565 PMCID: PMC7787192 DOI: 10.1016/j.cgh.2020.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts
General Hospital and Harvard Medical School, Boston, MA,Division of Gastroenterology, Massachusetts General
Hospital and Harvard Medical School, Boston, MA
| | - Lisa L. Strate
- Division of Gastroenterology, University of Washington
School of Medicine, Seattle, WA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts
General Hospital and Harvard Medical School, Boston, MA,Division of Gastroenterology, Massachusetts General
Hospital and Harvard Medical School, Boston, MA,Channing Division of Network Medicine, Department of
Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston,
MA,Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
22
|
Madison AA, Andridge R, Padin AC, Wilson S, Bailey MT, Alfano CM, Povoski SP, Lipari AM, Agnese DM, Carson WE, Malarkey WB, Kiecolt-Glaser JK. Endotoxemia coupled with heightened inflammation predicts future depressive symptoms. Psychoneuroendocrinology 2020; 122:104864. [PMID: 33166799 PMCID: PMC7721058 DOI: 10.1016/j.psyneuen.2020.104864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Cross-sectional data have linked gut barrier abnormalities and endotoxemia with depression, even among those without gastrointestinal symptoms. This study examined longitudinal associations between endotoxemia markers and depressive symptoms, as well as the role of inflammation in this relationship. DESIGN At three annual visits, 315 women (n=209 breast cancer survivors, n = 106 non-cancer patient controls, M=55 years old) completed the Center for Epidemiological Studies Depression questionnaire (CES-D) and provided blood samples to assess inflammatory markers - interleukin-6, tumor necrosis factor-alpha, and C-reactive protein - and endotoxemia markers - lipopolysaccharide-binding protein (LBP), soluble CD14 (sCD14), and their ratio. RESULTS Adjusting for key demographic variables, health behaviors, visit 1 depressive symptoms, and cancer status and treatment, women with higher visit 1 LBP and LBP/sCD14 had more depressive symptoms at the two subsequent annual visits. Illustrating the notable impact, a woman at the 75th percentile for LBP or LBP/sCD14 at visit 1 was 18 % more likely to report clinically significant depressive symptoms (CES-D ≥16) at follow-up than a woman in the lowest quartile. Cancer status and treatment type did not modulate this relationship. In contrast, visit 1 depressive symptoms did not predict endotoxemia at follow-up. A significant interaction between LBP/sCD14 and inflammatory burden suggested that visit 1 endotoxemia fueled depressive symptoms only in the context of elevated inflammation. CONCLUSION These results suggest that endotoxemia, combined with systemic inflammation, can drive depressive symptoms. These findings may implicate bacterial endotoxin translocation from the gut to the bloodstream in depression etiology. Interventions that reduce endotoxemia and inflammation may lessen the risk of depression.
Collapse
Affiliation(s)
- Annelise A. Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine,,Department of Psychology, The Ohio State University
| | - Rebecca Andridge
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine,,College of Public Health, The Ohio State University
| | - Avelina C. Padin
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine,,Department of Psychology, The Ohio State University
| | | | - Michael T. Bailey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine,,Department of Pediatrics, The Ohio State University College of Medicine,,Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | | | | | | | | | | | - William B. Malarkey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine,,Department of Internal Medicine, The Ohio State University College of Medicine
| | - Janice K. Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine,,Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine
| |
Collapse
|
23
|
Laugerette F, Vors C, Alligier M, Pineau G, Drai J, Knibbe C, Morio B, Lambert-Porcheron S, Laville M, Vidal H, Michalski MC. Postprandial Endotoxin Transporters LBP and sCD14 Differ in Obese vs. Overweight and Normal Weight Men during Fat-Rich Meal Digestion. Nutrients 2020; 12:nu12061820. [PMID: 32570947 PMCID: PMC7353369 DOI: 10.3390/nu12061820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
Circulating levels of lipopolysaccharide-binding protein (LBP) and soluble cluster of differentiation 14 (sCD14) are recognized as clinical markers of endotoxemia. In obese men, postprandial endotoxemia is modulated by the amount of fat ingested, being higher compared to normal-weight (NW) subjects. Relative variations of LBP/sCD14 ratio in response to overfeeding are also considered important in the inflammation set-up, as measured through IL-6 concentration. We tested the hypothesis that postprandial LBP and sCD14 circulating concentrations differed in obese vs. overweight and NW men after a fat-rich meal. We thus analyzed the postprandial kinetics of LBP and sCD14 in the context of two clinical trials involving postprandial tests in normal-, over-weight and obese men. In the first clinical trial eight NW and 8 obese men ingested breakfasts containing 10 vs. 40 g of fat. In the second clinical trial, 18 healthy men were overfed during 8 weeks. sCD14, LBP and Il-6 were measured in all subjects during 5 h after test meal. Obese men presented a higher fasting and postprandial LBP concentration in plasma than NW men regardless of fat load, while postprandial sCD14 was similar in both groups. Irrespective of the overfeeding treatment, we observed postprandial increase of sCD14 and decrease of LBP before and after OF. In obese individuals receiving a 10 g fat load, whereas IL-6 increased 5h after meal, LBP and sCD14 did not increase. No direct association between the postprandial kinetics of endotoxemia markers sCD14 and LBP and of inflammation in obese men was observed in this study.
Collapse
Affiliation(s)
- Fabienne Laugerette
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Correspondence: ; Tel.: +33-4-26-23-61-74
| | - Cécile Vors
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
| | - Maud Alligier
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310 Pierre-Bénite, France;
| | - Gaëlle Pineau
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
| | - Jocelyne Drai
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Laboratoire de Biochimie, Centre Hospitalier Lyon Sud, 69600 Oullins, France
| | - Carole Knibbe
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
| | - Béatrice Morio
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310 Pierre-Bénite, France;
| | - Stéphanie Lambert-Porcheron
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310 Pierre-Bénite, France;
- Hospices Civils de Lyon, 69000 Lyon, France
| | - Martine Laville
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310 Pierre-Bénite, France;
- Hospices Civils de Lyon, 69000 Lyon, France
| | - Hubert Vidal
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310 Pierre-Bénite, France;
- Hospices Civils de Lyon, 69000 Lyon, France
| | - Marie-Caroline Michalski
- Univ Lyon, CarMeN Laboratory, INRAE, UMR1397, INSERM, UMR1060, Université Claude Bernard Lyon 1, 69310 Pierre Bénite, France; (C.V.); (M.A.); (G.P.); (J.D.); (C.K.); (B.M.); (M.L.); (H.V.); (M.-C.M.)
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, 69310 Pierre-Bénite, France;
| |
Collapse
|
24
|
Roth RA, Ganey PE. What have we learned from animal models of idiosyncratic, drug-induced liver injury? Expert Opin Drug Metab Toxicol 2020; 16:475-491. [PMID: 32324077 DOI: 10.1080/17425255.2020.1760246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Idiosyncratic, drug-induced liver injury (IDILI) continues to plague patients and restrict the use of drugs that are pharmacologically effective. Mechanisms of IDILI are incompletely understood, and a better understanding would reduce speculation and could help to identify safer drug candidates preclinically. Animal models have the potential to enhance knowledge of mechanisms of IDILI. AREAS COVERED Numerous hypotheses have emerged to explain IDILI pathogenesis, many of which center on the roles of the innate and/or adaptive immune systems. Animal models based on these hypotheses are reviewed in the context of their contributions to understanding of IDILI and their limitations. EXPERT OPINION Animal models of IDILI based on an activated adaptive immune system have to date failed to reproduce major liver injury that is of most concern clinically. The only models that have so far resulted in pronounced liver injury are based on the multiple determinant hypothesis or the inflammatory stress hypothesis. The liver pathogenesis in IDILI animal models involves various leukocytes and immune mediators such as cytokines. Insights from animal models are changing the way we view IDILI pathogenesis and are leading to better approaches to preclinical prediction of IDILI potential of new drug candidates.
Collapse
Affiliation(s)
- Robert A Roth
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University , East Lansing, MI, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University , East Lansing, MI, USA
| |
Collapse
|
25
|
Abstract
Crohn's disease is an inflammatory bowel disease that is characterized by chronic inflammation of any part of the gastrointestinal tract, has a progressive and destructive course and is increasing in incidence worldwide. Several factors have been implicated in the cause of Crohn's disease, including a dysregulated immune system, an altered microbiota, genetic susceptibility and environmental factors, but the cause of the disease remains unknown. The onset of the disease at a young age in most cases necessitates prompt but long-term treatment to prevent disease flares and disease progression with intestinal complications. Thus, earlier, more aggressive treatment with biologic therapies or novel small molecules could profoundly change the natural history of the disease and decrease complications and the need for hospitalization and surgery. Although less invasive biomarkers are in development, diagnosis still relies on endoscopy and histological assessment of biopsy specimens. Crohn's disease is a complex disease, and treatment should be personalized to address the underlying pathogenetic mechanism. In the future, disease management might rely on severity scores that incorporate prognostic factors, bowel damage assessment and non-invasive close monitoring of disease activity to reduce the severity of complications.
Collapse
|
26
|
Moliterni E, Paolino G, Richetta AG, Grassi S, Calvieri S. The barrier beyond the skin. Ital J Dermatol Venerol 2019; 156:2-4. [PMID: 31804051 DOI: 10.23736/s2784-8671.19.06431-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Elisa Moliterni
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University, Rome, Italy -
| | | | - Antonio G Richetta
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University, Rome, Italy
| | - Sara Grassi
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University, Rome, Italy
| | - Stefano Calvieri
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University, Rome, Italy
| |
Collapse
|
27
|
Bian X, Wu W, Yang L, Lv L, Wang Q, Li Y, Ye J, Fang D, Wu J, Jiang X, Shi D, Li L. Administration of Akkermansia muciniphila Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Front Microbiol 2019; 10:2259. [PMID: 31632373 PMCID: PMC6779789 DOI: 10.3389/fmicb.2019.02259] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) develop as a result of complex interactions among genes, innate immunity and environmental factors, which are related to the gut microbiota. Multiple clinical and animal data have shown that Akkermansia muciniphila is associated with a healthy mucosa. However, its precise role in colitis is currently unknown. Our study aimed to determine its protective effects and underlying mechanisms in a dextran sulfate sodium (DSS)-induced colitis mouse model. Twenty-four C57BL/6 male mice were administered A. muciniphila MucT or phosphate-buffered saline (PBS) once daily by oral gavage for 14 days. Colitis was induced by drinking 2% DSS from days 0 to 6, followed by 2 days of drinking normal water. Mice were weighed daily and then sacrificed on day 8. We found that A. muciniphila improved DSS-induced colitis, which was evidenced by reduced weight loss, colon length shortening and histopathology scores and enhanced barrier function. Serum and tissue levels of inflammatory cytokines and chemokines (TNF-α, IL1α, IL6, IL12A, MIP-1A, G-CSF, and KC) decreased as a result of A. muciniphila administration. Analysis of 16S rDNA sequences showed that A. muciniphila induced significant gut microbiota alterations. Furthermore, correlation analysis indicated that pro-inflammatory cytokines and other injury factors were negatively associated with Verrucomicrobia, Akkermansia, Ruminococcaceae, and Rikenellaceae, which were prominently abundant in A. muciniphila-treated mice. We confirmed that A. muciniphila treatment could ameliorate mucosal inflammation either via microbe-host interactions, which protect the gut barrier function and reduce the levels of inflammatory cytokines, or by improving the microbial community. Our findings suggest that A. muciniphila may be a potential probiotic agent for ameliorating colitis.
Collapse
Affiliation(s)
- Xiaoyuan Bian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Wenrui Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Liya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Qing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Yating Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Jianzhong Ye
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Daiqiong Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Jingjing Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Xianwan Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| |
Collapse
|
28
|
Benz K, Schöbel A, Dietz M, Maurer P, Jackowski J. Adhesion Behaviour of Primary Human Osteoblasts and Fibroblasts on Polyether Ether Ketone Compared with Titanium under In Vitro Lipopolysaccharide Incubation. MATERIALS 2019; 12:ma12172739. [PMID: 31461861 PMCID: PMC6747843 DOI: 10.3390/ma12172739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022]
Abstract
The aim of this in vitro pilot study was to analyse the adhesion behaviour of human osteoblasts and fibroblasts on polyether ether ketone (PEEK) when compared with titanium surfaces in an inflammatory environment under lipopolysaccharide (LPS) incubation. Scanning electron microscopy (SEM) images of primary human osteoblasts/fibroblasts on titanium/PEEK samples were created. The gene expression of the LPS-binding protein (LBP) and the LPS receptor (toll-like receptor 4; TLR4) was measured by real-time polymerase chain reaction (PCR). Immunocytochemistry was used to obtain evidence for the distribution of LBP/TLR4 at the protein level of the extra-cellular-matrix-binding protein vinculin and the actin cytoskeleton. SEM images revealed that the osteoblasts and fibroblasts on the PEEK surfaces had adhesion characteristics comparable to those of titanium. The osteoblasts contracted under LPS incubation and a significantly increased LBP gene expression were detected. This was discernible at the protein level on all the materials. Whereas no increase of TLR4 was detected with regard to mRNA concentrations, a considerable increase in the antibody reaction was detected on all the materials. As is the case with titanium, the colonisation of human osteoblasts and fibroblasts on PEEK samples is possible under pro-inflammatory environmental conditions and the cellular inflammation behaviour towards PEEK is lower than that of titanium.
Collapse
Affiliation(s)
- Korbinian Benz
- Department of Oral Surgery and Dental Emergency Care, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany.
| | - Andreas Schöbel
- Department of Oral Surgery and Dental Emergency Care, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
| | - Marisa Dietz
- Department of Oral and Maxillofacial Surgery, Hospital North Dortmund, 44145 Dortmund, Germany
| | - Peter Maurer
- Private Practice Clinic for Oral Surgery, 66606 St. Wendel, Germany
| | - Jochen Jackowski
- Department of Oral Surgery and Dental Emergency Care, Faculty of Health, Witten/Herdecke University, 58455 Witten, Germany
| |
Collapse
|
29
|
Myers SA, Gobejishvili L, Saraswat Ohri S, Garrett Wilson C, Andres KR, Riegler AS, Donde H, Joshi-Barve S, Barve S, Whittemore SR. Following spinal cord injury, PDE4B drives an acute, local inflammatory response and a chronic, systemic response exacerbated by gut dysbiosis and endotoxemia. Neurobiol Dis 2018; 124:353-363. [PMID: 30557659 DOI: 10.1016/j.nbd.2018.12.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence links changes in the gut microbiome and intestinal barrier function to alterations in CNS function. We examined the role of endotoxin-responsive, cAMP-specific, Pde4 subfamily b (Pde4b) enzyme in gut dysbiosis induced neuro-inflammation and white matter loss following spinal cord injury (SCI). Using a thoracic contusion model in C57Bl/6 wild type female mice, SCI led to significant shifts in the gut bacterial community including an increase in the phylum Proteobacteria, which consists of endotoxin-harboring, gram-negative bacteria. This was accompanied by increased systemic inflammatory marker, soluble CD14, along with markers of the endoplasmic reticulum stress response (ERSR) and inflammation in the SCI epicenter. Deletion of Pde4b reduced epicenter expression of markers for the ERSR and inflammation, at both acute and chronic time points post-SCI. Correspondingly, expression of oligodendrocyte mRNAs increased. Within the injury penumbra, inflammatory protein markers of activated astrocytes (GFAP), macrophage/microglia (CD11b, Iba1), and the proinflammatory mediator Cox2, were decreased in Pde4b-/- mice. The absence of Pde4b improved white matter sparing and recovery of hindlimb locomotion following injury. Importantly, SCI-induced gut dysbiosis, bacterial overgrowth and endotoxemia were also prevented in Pde4b-/- mice. Taken together, these findings indicate that PDE4B plays an important role in the development of acute and chronic inflammatory response and consequent recovery following SCI.
Collapse
Affiliation(s)
- Scott A Myers
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA; Department of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Leila Gobejishvili
- Departments of Internal Medicine and Pharmacology and Toxicology, and Alcohol Research Center, University of Louisville, School of Medicine, 505 South Hancock Street, CTR Building, Room 515, Louisville, KY 40202, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA; Department of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - C Garrett Wilson
- UAB School of Medicine, University of Alabama at Birmingham, Bevill Biomedical Research Building, Birmingham, AL 35294, USA
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA; Department of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Amberly S Riegler
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA; Department of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Hridgandh Donde
- Departments of Internal Medicine and Pharmacology and Toxicology, and Alcohol Research Center, University of Louisville, School of Medicine, 505 South Hancock Street, CTR Building, Room 515, Louisville, KY 40202, USA
| | - Swati Joshi-Barve
- Departments of Internal Medicine and Pharmacology and Toxicology, and Alcohol Research Center, University of Louisville, School of Medicine, 505 South Hancock Street, CTR Building, Room 515, Louisville, KY 40202, USA
| | - Shirish Barve
- Departments of Internal Medicine and Pharmacology and Toxicology, and Alcohol Research Center, University of Louisville, School of Medicine, 505 South Hancock Street, CTR Building, Room 515, Louisville, KY 40202, USA.
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA; Department of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA; Department of Anatomical Science & Neurobiology, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA.
| |
Collapse
|
30
|
Fotis L, Shaikh N, Baszis KW, Samson CM, Lev-Tzion R, French AR, Tarr PI. Serologic Evidence of Gut-driven Systemic Inflammation in Juvenile Idiopathic Arthritis. J Rheumatol 2017; 44:1624-1631. [PMID: 28916545 PMCID: PMC5904838 DOI: 10.3899/jrheum.161589] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Accumulating evidence links juvenile idiopathic arthritis (JIA) to nonhost factors such as gut microbes. We hypothesize that children with new-onset JIA have increased intestinal bacterial translocation and circulating lipopolysaccharide (LPS). METHODS We studied systemic treatment-naive patients with JIA [polyarticular JIA, n = 22, oligoarticular JIA, n = 31, and spondyloarthropathies (SpA), n = 16], patients with established inflammatory bowel disease-related arthritis (IBD-RA, n = 11), and 34 healthy controls. We determined circulating IgG reactivity against LPS, LPS-binding protein (LBP), α-1-acid glycoprotein (α-1AGP), and C-reactive protein (CRP) in plasma or serum from these patients and controls. Juvenile Arthritis Disease Activity Score (JADAS-27) was calculated for patients with JIA. RESULTS Circulating anticore LPS antibody concentrations in patients with polyarticular JIA (p = 0.001), oligoarticular JIA (p = 0.024), and SpA (p = 0.001) were significantly greater than in controls, but there were no significant intergroup differences. Circulating LBP concentrations were also significantly greater in patients with polyarticular JIA (p = 0.001), oligoarticular JIA (p = 0.002), and SpA (p = 0.006) than controls, as were α-1AGP concentrations (p = 0.001, 0.001, and 0.003, respectively). No differences were observed between controls and patients with IBD-RA in any of the assays. Circulating concentrations of LBP and α-1AGP correlated strongly with CRP concentrations (r = 0.78 and r = 0.66, respectively). Anticore LPS antibody levels and CRP (r = 0.26), LBP (r = 0.24), and α-AGP (r = 0.22) concentrations had weaker correlations. JADAS-27 scores correlated with LBP (r = 0.66) and α-1AGP concentrations (r = 0.58). CONCLUSION Children with polyarticular JIA, oligoarticular JIA, and SpA have evidence of increased exposure to gut bacterial products. These data reinforce the concept that the intestine is a source of immune stimulation in JIA.
Collapse
Affiliation(s)
- Lampros Fotis
- Divisions of Rheumatology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
- Nottingham University Hospitals NHS Trust, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nurmohammad Shaikh
- Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Kevin W. Baszis
- Divisions of Rheumatology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Charles M. Samson
- Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Raffi Lev-Tzion
- Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
- Juliet Keidan Institute of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Anthony R. French
- Divisions of Rheumatology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Phillip I. Tarr
- Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
31
|
Dickerson F, Adamos M, Katsafanas E, Khushalani S, Origoni A, Savage C, Schweinfurth L, Stallings C, Sweeney K, Alaedini A, Uhde M, Severance E, Wilcox HC, Yolken R. The association between immune markers and recent suicide attempts in patients with serious mental illness: A pilot study. Psychiatry Res 2017; 255:8-12. [PMID: 28505469 DOI: 10.1016/j.psychres.2017.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 11/28/2022]
Abstract
Previous studies have identified elevations in markers of gastrointestinal inflammation in schizophrenia and mood disorders but studies have not measured the association between these markers and recent suicide attempts. We assessed 210 patients receiving treatment for schizophrenia, bipolar disorder, or major depression. We employed the Columbia Suicide Severity Rating Scale to identify recent and lifetime suicide attempts (actual, aborted, and interrupted). Psychiatric participants and a control group of 72 individuals without a psychiatric disorder had a blood sample drawn from which were measured specific markers of gastrointestinal inflammation and also C-Reactive protein (CRP). A total of 20 (10%) of psychiatric participants had a suicide attempt in the previous one month and 95 (45%) an attempt during their lifetime but not in the previous one month. The recent attempters had significantly elevated levels of antibodies to yeast mannan from Saccharomyces cerevisiae (ASCA), the food antigen gliadin, and bacterial lipopolysaccharide (LPS) compared with the non-psychiatric group when adjusting for demographic and clinical variables. These markers were not elevated in individuals with a past, but not recent, suicide attempt history. Our study indicates that there is evidence of gastrointestinal inflammation in some individuals who have had a recent suicide attempt.
Collapse
Affiliation(s)
- Faith Dickerson
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA.
| | - Maria Adamos
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Emily Katsafanas
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Sunil Khushalani
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Andrea Origoni
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Christina Savage
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Lucy Schweinfurth
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Cassie Stallings
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Kevin Sweeney
- Stanley Research Program, Sheppard Pratt Health System, Baltimore, MD, USA
| | - Armin Alaedini
- Department of Medicine, Columbia University, New York, NY, USA
| | - Melanie Uhde
- Department of Medicine, Columbia University, New York, NY, USA
| | - Emily Severance
- Stanley Neurovirology Laboratory, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Holly C Wilcox
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert Yolken
- Stanley Neurovirology Laboratory, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
32
|
Kwon JH, Im JP, Ye BD, Cheon JH, Jang HJ, Lee KM, Kim YS, Kim SW, Kim YH, Song GA, Han DS, Kim WH, Kim JS. Disease Phenotype, Activity and Clinical Course Prediction Based on C-Reactive Protein Levels at Diagnosis in Patients with Crohn's Disease: Results from the CONNECT Study. Gut Liver 2017; 10:595-603. [PMID: 27021506 PMCID: PMC4933421 DOI: 10.5009/gnl15411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/02/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIMS C-reactive protein (CRP) is an easily measured index of disease activity, but its ability to predict clinical course is controversial. We therefore designed a study to determine whether the CRP level at Crohn's disease (CD) diagnosis is a valuable indicator of the disease phenotype, activity, and clinical course. METHODS We retrospectively analyzed 705 CD patients from 32 institutions. The patients were classified into two groups according to CRP level. The patients' demographic and clinical characteristics and their use of immunosuppressive or biological agents were recorded. Disease location and behavior, hospitalization, and surgery were analyzed. RESULTS A high CRP was associated with younger age, steroid use, colonic or ileocolonic location, high CD activity index, and active inflammation at colonoscopy (p<0.001). As the disease progressed, patients with high CRP were more likely to exhibit strictures (p=0.027). There were significant differences in the use of 5-aminosalicylic acid, antibiotics, corticosteroids, azathioprine, and infliximab (p<0.001, p<0.001, p<0.001, p<0.001, and p=0.023, respectively). Hospitalization was also more frequent in patients with high CRP. CONCLUSIONS The CRP level at diagnosis is useful for evaluating the phenotype, activity, and clinical course of CD. Closer follow-up strategies, with early aggressive treatment, could be considered for patients with high CRP.
Collapse
Affiliation(s)
- Jee Hye Kwon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jong Pil Im
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Byong Duk Ye
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Joo Jang
- Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Kang Moon Lee
- Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea College of Medicine, Suwon, Korea
| | - You Sun Kim
- Department of Internal Medicine, Inje University College of Medicine, Seoul, Korea
| | - Sang Wook Kim
- Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Korea
| | - Young Ho Kim
- Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Geun Am Song
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Dong Soo Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, Korea
| | - Won Ho Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
33
|
Schmid W, Novacek G, Vogelsang H, Papay P, Primas C, Eser A, Panzer S. Platelets Toll-like receptor-4 in Crohns disease. Eur J Clin Invest 2017; 47:109-116. [PMID: 27714784 DOI: 10.1111/eci.12686] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Platelets are activated in Crohn's disease (CD) and interplay with leukocytes. Engagement of Toll-like receptor-4 (TLR-4), which is expressed in human platelets, may be involved in crosstalks between platelets and leukocytes leading to their mutual activation for host defense. MATERIALS AND METHODS Human neutrophil peptides (HNPs), lipoprotein binding peptides, and sCD14 were determined by enzyme-linked immunosorbent assays in 42 patients with active CD, in 43 patients with CD in remission, and in 30 healthy individuals. Neutrophil-platelet aggregates and binding of the TLR-4 monoclonal antibody to platelets were determined by flow cytometry. RESULTS Levels of HNPs were higher in patients with CD than in controls (P = 0.0003 vs. active CD and P = 0.01 vs. CD in remission). Likewise, neutrophils with adhering platelets were higher in patients with active CD than in controls (P = 0.004). Binding of the TLR-4 antibody in patients with active CD was similar to that in controls, while patients in remission had significantly higher binding capacities (P = 0.59 and P = 0.003). Incubation of plasma from patients with active disease or patients in remission with platelets from healthy controls confirmed lower binding of the TLR-4 antibody in the presence of plasma from active diseased patients compared to controls (P = 0.039), possibly due to high levels of lipopolysaccharides, as suggested by high levels of sCD14 and lipoprotein binding protein. CONCLUSION Our study indicates involvement of platelet TLR-4 in enhancing the secretion of antimicrobial peptides from neutrophils. While platelet aggregation can be due to a variety of mechanisms in inflammatory disease, the mutual activation of platelets and neutrophils may augment host defense.
Collapse
Affiliation(s)
- Werner Schmid
- Division of Cardiothoracic and Vascular Anesthesia and Intensive Care, Department of Anesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Gottfried Novacek
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Harald Vogelsang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Pavol Papay
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christian Primas
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Alexander Eser
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Simon Panzer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
Loganes C, Pin A, Naviglio S, Girardelli M, Bianco AM, Martelossi S, Tommasini A, Piscianz E. Altered pattern of tumor necrosis factor-alpha production in peripheral blood monocytes from Crohn's disease. World J Gastroenterol 2016; 22:9117-9126. [PMID: 27895399 PMCID: PMC5107593 DOI: 10.3748/wjg.v22.i41.9117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/25/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the inflammatory state in Crohn’s disease (CD) patients and correlate it with genetic background and microbial spreading.
METHODS By means of flow cytometry, production of tumor necrosis factor-alpha (TNF-α) was measured in peripheral blood monocytes from patients suffering from CD, ulcerative colitis (UC) and in healthy subjects after stimulation of the NOD2 and TLR pathways. CD patients were genotyped for the three most common NOD2 variants (R702W, G908R and L1007Pfs*2) and basal production of TNF-α was correlated to NOD2 genotype. Also, production of TNF-α was correlated to plasmatic levels of LPS Binding Protein (LBP), soluble (s) CD14 and to the activity state of the disease.
RESULTS The patients with CD were characterized by a significantly higher monocyte basal expression of TNF-α compared with healthy subjects and UC patients, and after stimulation with Pam3CSK4 (ligand of TLR2/1) and MDP-L18 (ligand of NOD2) this difference was maintained, while other microbial stimuli (LPS, ligand of TLR4 and PolyI:C, ligand of TLR3) induced massive activation in CD monocytes as well as in UC and in healthy control cells. There was no significant difference in the production of TNF-α between patients who carried CD-associated heterozygous or homozygous variants in NOD2 and patients with wild type NOD2 genotype. Although serum LBP levels have been shown to correlate positively with the state of activity of the disease, TNF-α production did not show a clear correlation with either LBP or sCD14 levels in plasma. Moreover, no clear correlation was seen between TNF-α production and activity indices in either CD or UC.
CONCLUSION Peripheral monocytes from CD express higher basal and stimulated TNF-α than controls, regardless of NOD2 genotype and without a clear correlation with disease activity.
Collapse
|
35
|
Citronberg JS, Wilkens LR, Lim U, Hullar MAJ, White E, Newcomb PA, Le Marchand L, Lampe JW. Reliability of plasma lipopolysaccharide-binding protein (LBP) from repeated measures in healthy adults. Cancer Causes Control 2016; 27:1163-6. [PMID: 27392432 DOI: 10.1007/s10552-016-0783-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/30/2016] [Indexed: 10/21/2022]
Abstract
Plasma lipopolysaccharide-binding protein (LBP), a measure of internal exposure to bacterial lipopolysaccharide, has been associated with several chronic conditions and may be a marker of chronic inflammation; however, no studies have examined the reliability of this biomarker in a healthy population. We examined the temporal reliability of LBP measured in archived samples from participants in two studies. In Study one, 60 healthy participants had blood drawn at two time points: baseline and follow-up (either three, six, or nine months). In Study two, 24 individuals had blood drawn three to four times over a seven-month period. We measured LBP in archived plasma by ELISA. Test-retest reliability was estimated by calculating the intraclass correlation coefficient (ICC). Plasma LBP concentrations showed moderate reliability in Study one (ICC 0.60, 95 % CI 0.43-0.75) and Study two (ICC 0.46, 95 % CI 0.26-0.69). Restricting the follow-up period improved reliability. In Study one, the reliability of LBP over a three-month period was 0.68 (95 % CI: 0.41-0.87). In Study two, the ICC of samples taken ≤seven days apart was 0.61 (95 % CI 0.29-0.86). Plasma LBP concentrations demonstrated moderate test-retest reliability in healthy individuals with reliability improving over a shorter follow-up period.
Collapse
Affiliation(s)
- Jessica S Citronberg
- Department of Epidemiology, University of Washington, Seattle, WA, USA. .,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, M4-B402, PO Box 19024, Seattle, WA, 98109, USA.
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Meredith A J Hullar
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, M4-B402, PO Box 19024, Seattle, WA, 98109, USA
| | - Emily White
- Department of Epidemiology, University of Washington, Seattle, WA, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, M4-B402, PO Box 19024, Seattle, WA, 98109, USA
| | - Polly A Newcomb
- Department of Epidemiology, University of Washington, Seattle, WA, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, M4-B402, PO Box 19024, Seattle, WA, 98109, USA
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Johanna W Lampe
- Department of Epidemiology, University of Washington, Seattle, WA, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, M4-B402, PO Box 19024, Seattle, WA, 98109, USA
| |
Collapse
|
36
|
Cardiolipins Act as a Selective Barrier to Toll-Like Receptor 4 Activation in the Intestine. Appl Environ Microbiol 2016; 82:4264-78. [PMID: 27208127 DOI: 10.1128/aem.00463-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/01/2016] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Intestinal homeostasis mechanisms must protect the host intestinal tissue from endogenous lipopolysaccharides (LPSs) produced by the intestinal microbiota. In this report, we demonstrate that murine intestinal fecal lipids effectively block Toll-like receptor 4 (TLR4) responses to naturally occurring Bacteroidetes sp. LPS. Cardiolipin (CL) represents a significant proportion of the total intestinal and fecal lipids and, furthermore, potently antagonizes TLR4 activation by reducing LPS binding at the lipopolysaccharide binding protein (LBP), CD14, and MD-2 steps of the TLR4 signaling pathway. It is further demonstrated that intestinal lipids and CL are less effective at neutralizing more potent Enterobacteriaceae-type LPS, which is enriched in feces obtained from mice with dextran sodium sulfate (DSS)-treated inflammatory bowel disease. The selective inhibition of naturally occurring LPS structures by intestinal lipids may represent a novel homeostasis mechanism that blocks LPS activation in response to symbiotic but not dysbiotic microbial communities. IMPORTANCE The guts of animals harbor a variety of Gram-negative bacteria associated with both states of intestinal health and states of disease. Environmental factors, such as dietary habits, can drive the microbial composition of the host animal's intestinal bacterial community toward a more pathogenic state. Both beneficial and harmful Gram-negative bacteria are capable of eliciting potentially damaging inflammatory responses from the host intestinal tissues via a lipopolysaccharide (LPS)-dependent pathway. Physical mucosal barriers and antibodies produced by the intestinal immune system protect against the undesired inflammatory effects of LPS, although it is unknown why some bacteria are more effective at overcoming the protective barriers than others. This report describes the discovery of a lipid-type protective barrier in the intestine that reduces the deleterious effects of LPSs from beneficial bacteria but is less effective in dampening the inflammatory effects of LPSs from harmful bacteria, providing a novel mechanistic insight into inflammatory intestinal disorders.
Collapse
|
37
|
Utility of surrogate markers for the prediction of relapses in inflammatory bowel diseases. J Gastroenterol 2016; 51:531-47. [PMID: 26975751 DOI: 10.1007/s00535-016-1191-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/21/2016] [Indexed: 02/04/2023]
Abstract
Patients with diagnosed inflammatory bowel disease (IBD) will commonly experience a clinical relapse in spite of a prolonged therapy-induced period of clinical remission. The current methods of assessing subclinical levels of low-grade inflammation which predispose patients to relapse are not optimal when considering both cost and patient comfort. Over the past few decades, much investigation has discovered that proteins such as calprotectin that are released from inflammatory cells are capable of indicating disease activity. Along with C-reactive protein and erythrocyte sedimentation rate, calprotectin has now become part of the current methodology for assessing IBD activity. More recently, research has identified that other fecal and serum biomarkers such as lactoferrin, S100A12, GM-CSF autoantibodies, α1-antitrypsin, eosinophil-derived proteins, and cytokine concentrations have variable degrees of utility in monitoring gastrointestinal tract inflammation. In order to provide direction toward novel methods of predicting relapse in IBD, we provide an up-to-date review of these biomarkers and their potential utility in the prediction of clinical relapse, given their observed activities during various stages of clinical remission.
Collapse
|
38
|
Zschemisch NH, Brüsch I, Hambusch AS, Bleich A. Transcription Factor SP2 Enhanced the Expression of Cd14 in Colitis-Susceptible C3H/HeJBir. PLoS One 2016; 11:e0155821. [PMID: 27191968 PMCID: PMC4871554 DOI: 10.1371/journal.pone.0155821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/04/2016] [Indexed: 01/19/2023] Open
Abstract
Genetic analysis in the IL10-deficient mouse model revealed a modifier locus of experimental inflammatory bowel disease (IBD) on chromosome 18, with the allele of the strain C3H/HeJBir (C3Bir) conferring resistance and the allele of C57BL/6J (B6) conferring susceptibility. Differential Cd14 expression was associated with this background specific susceptibility to intestinal inflammation. Polymorphisms of the Cd14 promoter were found to be likely causative for strain specific expression, and Cd14-knockout mice revealed a protective role of this gene-product in experimental IBD. In this study, luciferase reporter assays confirmed an increased activity of the C3Bir derived Cd14 promoter compared to the one of B6. Promoter truncation experiments and site-directed mutagenesis in both strains resulted in reduced Cd14 promoter activity and confirmed that a central AP1 and the proximal SP1 transcription factor binding sites mediated the basal activity of the Cd14 promoter in the mouse. Moreover, a T to C exchange at position -259 replaced putative STAT1 and CDX1 sites in the Cd14 promoter from B6 by a SP2 site in C3Bir. Ablation of the Sp2 site through truncation was associated with a decreased promoter activity. Site-directed mutagenesis also demonstrated that the inactivation of SP2 led to a substantial loss of promoter activity in C3Bir. Performing electrophoretic mobility shift and supershift assays demonstrated interaction of SP2 with its potential binding site. In addition, retroviral—mediated overexpression of the SP2 transcription factor in primary bone marrow macrophages derived from C3Bir mice caused a significant increase in Cd14 transcription. These data characterized SP2 as important factor responsible for higher Cd14 expression and reduced IBD susceptibility mediated by the C3Bir allele.
Collapse
Affiliation(s)
- Nils-Holger Zschemisch
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Inga Brüsch
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Anne-Sophie Hambusch
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
39
|
Dullah EC, Ongkudon CM. Current trends in endotoxin detection and analysis of endotoxin–protein interactions. Crit Rev Biotechnol 2016; 37:251-261. [DOI: 10.3109/07388551.2016.1141393] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Elvina Clarie Dullah
- Biotechnology Research Institute, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Clarence M. Ongkudon
- Biotechnology Research Institute, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
40
|
Gnauck A, Lentle RG, Kruger MC. Chasing a ghost?--Issues with the determination of circulating levels of endotoxin in human blood. Crit Rev Clin Lab Sci 2016; 53:197-215. [PMID: 26732012 DOI: 10.3109/10408363.2015.1123215] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reliable quantification of bacterial products such as endotoxin is important for the diagnosis of Gram-negative infection and for the monitoring of its treatment. Further, it is important to identify patients with persistent subclinical level of bacterial products in their systemic circulation as data from animal studies also suggest this may be correlated with the onset of metabolic syndrome. In this review, we first aim to describe the principles of the Limulus amoebocyte lysate (LAL) test, an assay that is used to quantify endotoxin, and the various shortcomings that must be addressed before it can become a reliable means of quantifying endotoxin in samples derived from blood. We then review published data regarding endotoxin levels in healthy subjects and those with sepsis, inflammatory bowel disease, liver disorders and metabolic disorders such as obesity and diabetes. We also review the evidence regarding influence of macronutrients in augmenting the levels of systemic endotoxin. The results of this review show that reported mean levels of endotoxin in the systemic circulation of healthy humans and of those with various clinical disorders vary over a wide range. Further, this review shows that a significant proportion of this variation can be related to the method that was used to prepare plasma and serum samples prior to assay and its ability to reduce the effect of various blood borne factors that interfere with the LAL assay.
Collapse
Affiliation(s)
- Anne Gnauck
- a Physiology Group, School of Food and Nutrition, College of Health, Massey University , Palmerston North , New Zealand
| | - Roger Graham Lentle
- a Physiology Group, School of Food and Nutrition, College of Health, Massey University , Palmerston North , New Zealand
| | - Marlena Cathorina Kruger
- a Physiology Group, School of Food and Nutrition, College of Health, Massey University , Palmerston North , New Zealand
| |
Collapse
|
41
|
Dlugosz A, Nowak P, D'Amato M, Mohammadian Kermani G, Nyström J, Abdurahman S, Lindberg G. Increased serum levels of lipopolysaccharide and antiflagellin antibodies in patients with diarrhea-predominant irritable bowel syndrome. Neurogastroenterol Motil 2015; 27:1747-54. [PMID: 26387872 DOI: 10.1111/nmo.12670] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/11/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Innate immune responses to conserved microbial products such as lipopolysaccharide (LPS) and flagellin are likely important in microbial-host interactions and intestinal homeostasis. We hypothesized that bacterial translocation and activation of mucosal immunity against common microbial antigens might be involved in the development of irritable bowel syndrome (IBS). We therefore compared serum levels of LPS, soluble CD14 (sCD14), and flagellin antibodies between patients with different subtypes of IBS and healthy controls. METHODS We analyzed serum obtained from 88 patients (74 females) aged 19(43)-73 years and 106 healthy volunteers (77 females) aged 19(38)-62 years. Diarrhea-predominant IBS (D-IBS) was present in 32 patients (36%), 23 patients (26%) had constipation-predominant IBS (C-IBS), and 33 patients (38%) had A-IBS. We used ELISA for sCD14 and antiflagellin immunoglobulin G and limulus amebocyte assay for LPS. Abdominal symptoms and psychiatric comorbidities were assessed using validated questionnaires. KEY RESULTS We found a significantly higher serum level of LPS in patients with D-IBS compared to controls (p = 0.0155). The level of antibodies to flagellin was higher in patients with IBS than in controls (mainly driven by higher levels in D-IBS, p = 0.0018). The levels of sCD14 were lower in D-IBS patients compared to controls (p = 0.0498). We found a weak, but significant correlation between the levels of antiflagellin antibodies and anxiety among IBS patients (ρ = 0.38; p = 0.0045). CONCLUSIONS & INFERENCES Our results support the concept that immune reactivity to luminal antigens may have a role in the development of D-IBS. The serum level of antiflagellin antibodies was found to correlate with patients' self-reported anxiety score.
Collapse
Affiliation(s)
- A Dlugosz
- Karolinska Institutet, Department of Medicine and Center for Digestive Diseases, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - P Nowak
- Karolinska Institutet, Department of Medicine, Unit of Infectious Diseases, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - M D'Amato
- Karolinska Institutet, Department of Biosciences and Nutrition, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - G Mohammadian Kermani
- Karolinska Institutet, Department of Medicine and Center for Digestive Diseases, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - J Nyström
- Karolinska Institutet, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - S Abdurahman
- Department of Science and Technology, Örebro Life Science Center, Örebro University, Örebro, Sweden
| | - G Lindberg
- Karolinska Institutet, Department of Medicine and Center for Digestive Diseases, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
42
|
Ben Ya'acov A, Lichtenstein Y, Zolotarov L, Ilan Y. The gut microbiome as a target for regulatory T cell-based immunotherapy: induction of regulatory lymphocytes by oral administration of anti-LPS enriched colostrum alleviates immune mediated colitis. BMC Gastroenterol 2015; 15:154. [PMID: 26518263 PMCID: PMC4628342 DOI: 10.1186/s12876-015-0388-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/25/2015] [Indexed: 02/07/2023] Open
Abstract
Background Gut-derived bacterial endotoxin is an important cofactor in the pathogenesis of IBD. Regulatory T cells (Tregs) are essential for maintenance of peripheral tolerance and can prevent and alleviate IBD. To determine the immune modulatory effect of anti-LPS enriched hyperimmune colostrum, its ability to induce Tregs and alleviate immune mediated colitis. Methods Immune-mediated colitis was induced in mice by intra-colonic instillation of Trinitrobenzene Sulfonate (TNBS). Four groups of mice were orally administered with two dosages of IgG-enriched colostrum fractions. The fractions were harvested from cows immunized against LPS derived from intestinal Escherichia coli bacteria (Imm124E). Control mice received non-immunized colostrum or vehicle (PBS). Treatment was administered one day following sensitization and four additional days following the administration of TNBS. The following parameters in the mice were tracked: body weight, bowel histology, serum cytokine levels and regulatory T cells. Results Oral administration of Imm124E hyperimmune colostrum ameliorated immune-mediated colitis. Significant amelioration of weight reduction was noted in treated mice. Oral administration of Imm124E improved bowel histology. Both the extent of the disease, inflammation score, and colitis damage and regeneration scores decreased in Imm-124E treated animals. These effects were associated with an increase in serum IL10 anti inflammatory cytokine levels, and an increase in CD4 + CD25+ and CD4 + Foxp3+ Tregs. Conclusions Oral administration of Imm124E promoted Tregs and alleviated bowel inflammation in immune mediated colitis. The present data suggests that the microbiome may serve as a target for Tregs-based immunotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s12876-015-0388-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ami Ben Ya'acov
- Gastroenterology and liver Unit, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| | - Yoav Lichtenstein
- Gastroenterology and liver Unit, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| | - Lidya Zolotarov
- Gastroenterology and liver Unit, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| | - Yaron Ilan
- Gastroenterology and liver Unit, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
43
|
Maxwell JR, Zhang Y, Brown WA, Smith CL, Byrne FR, Fiorino M, Stevens E, Bigler J, Davis JA, Rottman JB, Budelsky AL, Symons A, Towne JE. Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity 2015; 43:739-50. [PMID: 26431947 DOI: 10.1016/j.immuni.2015.08.019] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/28/2015] [Accepted: 08/20/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-23 (IL-23) and IL-17 are cytokines currently being targeted in clinical trials. Although inhibition of both of these cytokines is effective for treating psoriasis, IL-12 and IL-23 p40 inhibition attenuates Crohn's disease, whereas IL-17A or IL-17 receptor A (IL-17RA) inhibition exacerbates Crohn's disease. This dichotomy between IL-23 and IL-17 was effectively modeled in the multidrug resistance-1a-ablated (Abcb1a(-/-)) mouse model of colitis. IL-23 inhibition attenuated disease by decreasing colonic inflammation while enhancing regulatory T (Treg) cell accumulation. Exacerbation of colitis by IL-17A or IL-17RA inhibition was associated with severe weakening of the intestinal epithelial barrier, culminating in increased colonic inflammation and accelerated mortality. These data show that IL-17A acts on intestinal epithelium to promote barrier function and provide insight into mechanisms underlying exacerbation of Crohn's disease when IL-17A or IL-17RA is inhibited.
Collapse
Affiliation(s)
- Joseph R Maxwell
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - Yu Zhang
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - William A Brown
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - Carole L Smith
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - Fergus R Byrne
- Department of Inflammation, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Mike Fiorino
- Department of Inflammation, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Erin Stevens
- Department of Clinical Immunology, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - Jeannette Bigler
- Department of Molecular Sciences and Computational Biology, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - John A Davis
- Department of Pharmacokinetics and Drug Metabolism, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - James B Rottman
- Department of Pathology, Amgen, Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Alison L Budelsky
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - Antony Symons
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA
| | - Jennifer E Towne
- Department of Inflammation, Amgen, Inc., 1201 Amgen Court West, Seattle, WA 98117, USA.
| |
Collapse
|
44
|
Vesterbacka J, Barqasho B, Häggblom A, Nowak P. Effects of Co-Trimoxazole on Microbial Translocation in HIV-1-Infected Patients Initiating Antiretroviral Therapy. AIDS Res Hum Retroviruses 2015; 31:830-6. [PMID: 26059763 DOI: 10.1089/aid.2014.0366] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microbial translocation (MT) contributes to immune activation during HIV-1 infection, and persists after initiation of antiretroviral therapy (ART). We investigated whether levels of MT markers are influenced by the use of co-trimoxazole (TMP-SMX) in HIV-1 patients. Plasma samples were obtained from HIV-1-infected patients initiating ART with (n=13) or without (n=13) TMP-SMX prophylaxis. Markers of MT [lipopolysaccharide-binding protein (LBP), lipopolysaccharide (LPS), soluble CD14 (sCD14), and intestinal fatty acid binding protein (I-FABP)] were assessed at baseline (BL), at 1 month, and at 1 year by the Limulus Amebocyte Lysate Assay or ELISA. BL levels of LBP were elevated in both categories of patients; they were highest in patients starting ART and TMP-SMX (median, μg/ml: 36.7 vs. 4.3, respectively, p=0.001) and correlated inversely with CD4(+) T cell counts (ρ=-0.65; p=0.005). Patients receiving ART and TMP-SMX had a significant reduction in LBP between BL and 1 year (median, μg/ml: 36.7 vs. 11.1; p=0.003). In contrast, levels of LPS at BL were lower in patients starting ART and TMP-SMX compared to those without TMP-SMX (median, pg/ml: 221 vs. 303 respectively; p=0.002) and did not change at 1 year. The increased BL levels of sCD14 had declined in both groups at 1 year. No difference in I-FABP levels was found between BL and 1 year. Concomitant use of ART and TMP-SMX reduces microbial translocation markers LBP and sCD14, probably due to its impact on the gut microbiota. Effective ART for 1 year does not restore gut-blood barrier dysfunction.
Collapse
Affiliation(s)
- Jan Vesterbacka
- Department of Medicine, Unit of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Babilonia Barqasho
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Amanda Häggblom
- Department of Medicine, Unit of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Infectious Diseases, County Council of Gävleborg, Gävle, Sweden
| | - Piotr Nowak
- Department of Medicine, Unit of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
45
|
Fung TC, Artis D, Sonnenberg GF. Anatomical localization of commensal bacteria in immune cell homeostasis and disease. Immunol Rev 2015; 260:35-49. [PMID: 24942680 DOI: 10.1111/imr.12186] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mammalian gastrointestinal (GI) tract is colonized by trillions of beneficial commensal bacteria that are essential for promoting normal intestinal physiology. While the majority of commensal bacteria are found in the intestinal lumen, many species have also adapted to colonize different anatomical locations in the intestine, including the surface of intestinal epithelial cells (IECs) and the interior of gut-associated lymphoid tissues. These distinct tissue localization patterns permit unique interactions with the mammalian immune system and collectively influence intestinal immune cell homeostasis. Conversely, dysregulated localization of commensal bacteria can lead to inappropriate activation of the immune system and is associated with numerous chronic infectious, inflammatory, and metabolic diseases. Therefore, regulatory mechanisms that control proper anatomical containment of commensal bacteria are essential to maintain tissue homeostasis and limit pathology. In this review, we propose that commensal bacteria associated with the mammalian GI tract can be anatomically defined as (i) luminal, (ii) epithelial-associated, or (iii) lymphoid tissue-resident, and we discuss the role and regulation of these microbial populations in health and disease.
Collapse
Affiliation(s)
- Thomas C Fung
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
46
|
Ghigliotti G, Barisione C, Garibaldi S, Fabbi P, Brunelli C, Spallarossa P, Altieri P, Rosa G, Spinella G, Palombo D, Arsenescu R, Arsenescu V. Adipose tissue immune response: novel triggers and consequences for chronic inflammatory conditions. Inflammation 2014; 37:1337-53. [PMID: 24823865 PMCID: PMC4077305 DOI: 10.1007/s10753-014-9914-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adipose tissue inflammation mediates the association between excessive body fat accumulation and several chronic inflammatory diseases. A high prevalence of obesity-associated adipose tissue inflammation was observed not only in patients with cardiovascular conditions but also in patients with inflammatory bowel diseases, abdominal aortic aneurysm, or cardiorenal syndrome. In addition to excessive caloric intake, other triggers promote visceral adipose tissue inflammation followed by chronic, low-grade systemic inflammation. The infiltration and accumulation of immune cells in the inflamed and hypertrophied adipose tissue promote the production of inflammatory cytokines, contributing to target organ damages. This comorbidity seems to delimit subgroups of individuals with systemic adipose tissue inflammation and more severe chronic inflammatory diseases that are refractory to conventional treatment. This review highlights the association between adipose tissue immune response and the pathophysiology of visceral adiposity-related chronic inflammatory diseases, while suggesting several new therapeutic strategies.
Collapse
Affiliation(s)
- Giorgio Ghigliotti
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Chiara Barisione
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Silvano Garibaldi
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Patrizia Fabbi
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Claudio Brunelli
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Paolo Spallarossa
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Paola Altieri
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Gianmarco Rosa
- Division of Cardiology, IRCCS University Hospital San Martino, Research Centre of Cardiovascular Biology, University of Genoa, Genoa, Italy
| | - Giovanni Spinella
- Vascular and Endovascular Surgery Unit, University of Genoa, Genoa, Italy
| | - Domenico Palombo
- Vascular and Endovascular Surgery Unit, University of Genoa, Genoa, Italy
| | - Razvan Arsenescu
- IBD Center, Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, Columbus, OH USA
| | - Violeta Arsenescu
- Mucosal Immunology IBD Laboratory, Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University, 400W 12 Ave., Wiseman Hall, Room 1024, Columbus, OH 43210 USA
| |
Collapse
|
47
|
Biomarkers of inflammatory bowel disease. DISEASE MARKERS 2014; 2014:710915. [PMID: 24963213 PMCID: PMC4055235 DOI: 10.1155/2014/710915] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/06/2014] [Indexed: 12/13/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic disease mostly involved with intestine with unknown etiology. Diagnosis, evaluation of severity, and prognosis are still present as challenges for physicians. An ideal biomarker with the characters such as simple, easy to perform, noninvasive or microinvasive, cheap, rapid, and reproducible is helpful for patients and clinicians. Currently biomarkers applied in clinic include CRP, ESR, pANCA, ASCA, and fecal calprotectin. However, they are far from ideal. Lots of studies are focused on seeking for ideal biomarker for IBD. Herein, the paper reviewed recent researches on biomarkers of IBD to get advances of biomarkers in inflammatory bowel disease.
Collapse
|
48
|
Magro F, Sousa P, Ministro P. C-reactive protein in Crohn's disease: how informative is it? Expert Rev Gastroenterol Hepatol 2014; 8:393-408. [PMID: 24635486 DOI: 10.1586/17474124.2014.893821] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
C-reactive protein (CRP) is an important acute-phase marker, produced mainly in the liver. Its production by mesenteric adipocytes has been recently stressed in Crohn's disease (CD). There are many factors affecting CRP levels, both environmental and genetics. The short-life of this biomarker makes it of pertinent use in the assessment of inflammation. There are inconsistent results concerning the association of clinical activity indices, mucosal healing, histological activity and CRP. This review summarizes the role of CRP in CD, namely its importance in the differential diagnosis of CD; its relationship with clinical activity indices, other markers of inflammation and endoscopic and radiological cross sectional imaging; prediction of response to anti-TNF treatment and prediction of outcome.
Collapse
Affiliation(s)
- Fernando Magro
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Alameda Prof. Hernani Monteiro, 420-319 Porto, Portugal
| | | | | |
Collapse
|
49
|
Ding PH, Jin L. The role of lipopolysaccharide-binding protein in innate immunity: a revisit and its relevance to oral/periodontal health. J Periodontal Res 2013; 49:1-9. [DOI: 10.1111/jre.12081] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2013] [Indexed: 12/28/2022]
Affiliation(s)
- P.-H. Ding
- Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| | - L.J. Jin
- Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| |
Collapse
|
50
|
Abstract
At mucosal surfaces, phagocytes such as macrophages coexist with microbial communities; highly controlled regulation of these interactions is essential for immune homeostasis. Pattern-recognition receptors (PRRs) are critical in recognizing and responding to microbial products, and they are subject to negative regulation through various mechanisms, including downregulation of PRR-activating components or induction of inhibitors. Insights into these regulatory mechanisms have been gained through human genetic disease-association studies, in vivo mouse studies utilizing disease models or targeted gene perturbations, and in vitro and ex vivo human cellular studies examining phagocytic cell functions. Although mouse models provide an important approach to study macrophage regulation, human and mouse macrophages exhibit differences, which must be considered when extrapolating mouse findings to human physiology. This review discusses inhibitory regulation of PRR-induced macrophage functions and the consequences of dysregulation of these functions and highlights mechanisms that have a role in intestinal macrophages and in human macrophage studies.
Collapse
Affiliation(s)
- M Hedl
- Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| | | |
Collapse
|