1
|
Mathew AM, Huber A, Sous RD, Weghorn KN, Powers-Fletcher MV, Jose S, Madan R. Effect of Leptin Receptor Q223R Polymorphism on Clostridioides difficile Infection-Induced Macrophage Migration Inhibitory Factor Production. J Infect Dis 2024; 230:816-820. [PMID: 38687212 PMCID: PMC11481448 DOI: 10.1093/infdis/jiae141] [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: 11/08/2023] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Proinflammatory cytokine levels and host genetic makeup are key determinants of Clostridioides difficile infection (CDI) outcomes. We previously reported that blocking the inflammatory cytokine macrophage migration inhibitory factor (MIF) ameliorates CDI. Here, we determined kinetics of MIF production and its association with a common genetic variant in leptin receptor (LEPR) using blood from patients with CDI. We found highest plasma MIF early after C difficile exposure and in individuals who express mutant/derived LEPR. Our data suggest that early-phase CDI provides a possible window of opportunity in which MIF targeting, potentially in combination with LEPR genotype, could have therapeutic utility.
Collapse
Affiliation(s)
- Ann M Mathew
- Division of Infectious Diseases
- Pathobiology and Molecular Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine
| | - Alexander Huber
- Division of Infectious Diseases
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center
| | | | | | - Margaret V Powers-Fletcher
- Division of Infectious Diseases
- Pathobiology and Molecular Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine
| | | | - Rajat Madan
- Division of Infectious Diseases
- Pathobiology and Molecular Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center
- Veterans Affairs Medical Center Cincinnati, Cincinnati, Ohio
| |
Collapse
|
2
|
Huang J, Liu B, Liu Y, You W, Zhao P, Liu Y, Wang K, Lai X, Xu B, Cao J. Progranulin protects against Clostridioides difficile infection by enhancing IL-22 production. Gut Microbes 2024; 16:2409220. [PMID: 39349385 PMCID: PMC11444516 DOI: 10.1080/19490976.2024.2409220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/02/2024] Open
Abstract
Enhanced mortality, relapse rates, and increased prevalence of Clostridioides difficile infection (CDI) emphasize the need for better therapies and management approaches. Modulating host immune response to ameliorate CDI-associated immunopathology may provide new advantages to currently inadequate antibiotic therapies. Here, we identified progranulin (PGRN) as an important immune target upregulated in response to CDI. PGRN-deficient mice displayed dramatically higher mortality and aggravated epithelial barrier disruption compared with wild type (WT) mice after CDI despite equivalent levels of bacterial burden or toxin in the large intestine. Mechanistically, PGRN protection was mediated by IL-22 production from CD4+ T helper cells, as demonstrated by a decrease in colonic IL-22-producing CD4+ T helper cells in the intestine of PGRN-deficient mice upon CDI and a boost of IL-22-producing CD4+ T helper cells activated by PGRN ex vivo. Clinical evidence suggests that CDI patients had significantly higher serum levels of PGRN compared with healthy controls, which was significantly and positively correlated with IL-22. Our findings thus indicate a critical role for PGRN-promoted CD4+ T cell IL-22 production in shaping gut immunity and reestablishing the intestinal barrier during CDI. As an alternative to pathogen-targeted therapy, this study may provide a new host-directed therapeutic strategy to attenuate severe, refractory CDI.
Collapse
Affiliation(s)
- Jun Huang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bichen Liu
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yi Liu
- Department of Surgery, School of Medicine, Stanford University, Stanford, USA
| | - Wenxian You
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuhan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kehan Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofei Lai
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Muroya D, Nadayoshi S, Yamada K, Kai Y, Masuda N, Nishida T, Shimokobe M, Hisaka T. Effects of Hyperbaric Oxygen Therapy for Clostridioides difficile-associated Colitis: A Retrospective Study. J Anus Rectum Colon 2023; 7:264-272. [PMID: 37900689 PMCID: PMC10600261 DOI: 10.23922/jarc.2023-033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/20/2023] [Indexed: 10/31/2023] Open
Abstract
Objectives Clostridioides difficile (CD) is an anaerobic spore-forming Gram-positive rod that is a major cause of antibiotic-associated diarrhea. Hyperbaric oxygen therapy (HBO) is a well-established treatment for Clostridium perfringens, but there are no reports that have examined the efficacy of HBO against CD, which is also an anaerobic bacterium. Methods In this study, we retrospectively examined whether HBO therapy affects the prognosis following CD infections (CDI). This study included 92 inpatients diagnosed with CDI at our hospital between January 2013 and December 2022. Of these, 16 patients received HBO therapy. The indications for HBO therapy were stroke in five patients, ileus in four patients, cancer in two patients, acute peripheral circulatory disturbance in two patients, and others in three patients. The mean observation period was 5.4 years. Results In the univariate analysis, there was no significant difference in severity, mortality, hospitalization, or overall survival between patients who did and did not receive HBO therapy. However, the HBO group had a significantly lower recurrence rate (0% vs. 22.4%, p=0.0363) and a shorter symptomatic period (6.2 vs. 13.6 days, p=0.0217). Conclusions HBO may have beneficial effect on CDI by shortening the symptomatic period and preventing recurrence.
Collapse
Affiliation(s)
- Daisuke Muroya
- Department of Surgery, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Shinya Nadayoshi
- Department of Clinical Engineering, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
| | - Koito Yamada
- Department of Clinical Engineering, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
| | - Yutaro Kai
- Department of Clinical Engineering, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
| | - Naoki Masuda
- Department of Surgery, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
| | - Takamichi Nishida
- Department of Internal Medicine, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
| | - Masayuki Shimokobe
- Department of Internal Medicine, Tobata Kyoritsu Hospital, Kitakyusyu, Japan
| | - Toru Hisaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
4
|
Boicean LC, Birlutiu RM, Birlutiu V. Correlations between serum leptin levels and classical biomarkers in SARS-CoV-2 infection, in critically ill patients. Microb Pathog 2023; 182:106238. [PMID: 37419217 DOI: 10.1016/j.micpath.2023.106238] [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: 11/03/2021] [Revised: 04/30/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
BACKGROUND Altered levels of some blood markers might be linked with the degree of severity and mortality of patients with SARS-CoV-2 infection. This study aimed to find out if there are correlations between serum leptin levels and classical biomarkers. MATERIALS AND METHODS We present a single-center observational cohort study on SARS-CoV-2 infected patients. The study was conducted at Infectious Diseases Clinic of Academic Emergency Hospital Sibiu, from May through November 2020. In this study, we retrospectively analyzed 54 patients, all with confirmed SARS-CoV-2 infection. RESULTS Our results revealed that there is a negative correlation between serum leptin and Interleukin-6 levels and a positive correlation between serum leptin and blood glucose levels. A positive correlation between ferritin and lactate dehydrogenase levels was also observed. No correlation was found between leptin and other biomarkers such as ferritin, neutrophil/lymphocyte ratio, lactate dehydrogenase, C-reactive protein, fibrinogen, erythrocyte sedimentation rate, or D-dimer. CONCLUSIONS Further studies need to be conducted to investigate the role of leptin in SARS-CoV-2 infection. The results of this research could contribute to the introduction of the determination of serum leptin levels in the routine evaluation of patients with critical illness.
Collapse
Affiliation(s)
- Loredana Camelia Boicean
- "Lucian Blaga" University of Sibiu, Faculty of Medicine, Sibiu, Romania; Academic Emergency Hospital Sibiu, Infectious Diseases Clinic, Sibiu, Romania.
| | | | - Victoria Birlutiu
- "Lucian Blaga" University of Sibiu, Faculty of Medicine, Sibiu, Romania; Academic Emergency Hospital Sibiu, Infectious Diseases Clinic, Sibiu, Romania
| |
Collapse
|
5
|
Malick A, Wang Y, Axelrad J, Salmasian H, Freedberg D. Obesity is not associated with adverse outcomes among hospitalized patients with Clostridioides difficile infection. Gut Pathog 2022; 14:7. [PMID: 35093158 PMCID: PMC8799984 DOI: 10.1186/s13099-022-00479-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/06/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Obesity is associated with increased risk for death in most infections but has not been studied as a risk factor for mortality in Clostridioides difficile infection (CDI). This study tested obesity as a risk factor for death in patients hospitalized with CDI. This was a three-center retrospective study that included hospitalized adults with CDI at Columbia University Irving Medical Center, Brigham and Women’s Hospital, and NYU Langone from 2010 to 2018. Multivariate logistic regression was used to assess the relationship between obesity, measured by body mass index, and death from any cause within 30 days after the index CDI test.
Results
Data for 3851 patients were analyzed, including 373 (9.7%) who died within 30 days following a diagnosis of CDI. After adjusting for other factors, BMI was not associated with increased risk for death in any BMI category [adjusted OR (aOR) 0.96, 95% CI 0.69 to 1.34 for BMI > 30 vs BMI 20–30; aOR 1.02, 95% CI 0.53 to 1.87 for BMI > 40 vs BMI 20–30]. After stratifying into three groups by age, there were trends towards increased mortality with obesity in the middle-aged (56–75 vs ≤ 55 years old) yet decreased mortality with obesity in the old (> 75 vs ≤ 55) (p = NS for all). Advanced age and low albumin were the factors most strongly associated with death.
Conclusions
We found no association between obesity and death among patients with CDI, in contrast to most other infections. Obesity is not likely to be useful for risk-stratifying hospitalized patients with CDI.
Collapse
|
6
|
Chandra H, Sharma KK, Tuovinen OH, Sun X, Shukla P. Pathobionts: mechanisms of survival, expansion, and interaction with host with a focus on Clostridioides difficile. Gut Microbes 2022; 13:1979882. [PMID: 34724858 PMCID: PMC8565823 DOI: 10.1080/19490976.2021.1979882] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Pathobionts are opportunistic microbes that emerge as a result of perturbations in the healthy microbiome due to complex interactions of various genetic, exposomal, microbial, and host factors that lead to their selection and expansion. Their proliferations can aggravate inflammatory manifestations, trigger autoimmune diseases, and lead to severe life-threatening conditions. Current surge in microbiome research is unwinding these complex interplays between disease development and protection against pathobionts. This review summarizes the current knowledge of pathobiont emergence with a focus on Clostridioides difficile and the recent findings on the roles of immune cells such as iTreg cells, Th17 cells, innate lymphoid cells, and cytokines in protection against pathobionts. The review calls for adoption of innovative tools and cutting-edge technologies in clinical diagnostics and therapeutics to provide insights in identification and quantification of pathobionts.
Collapse
Affiliation(s)
- Harish Chandra
- Department of Environmental Microbiology, School of Earth and Environmental Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India,Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Krishna Kant Sharma
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Olli H. Tuovinen
- Department of Microbiology, Ohio State University, Columbus, OH, USA
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA,Xingmin Sun Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Pratyoosh Shukla
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India,Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, India,CONTACT Pratyoosh Shukla School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
7
|
Mohammadi MR, Rodriguez SM, Luong JC, Li S, Cao R, Alshetaiwi H, Lau H, Davtyan H, Jones MB, Jafari M, Kessenbrock K, Villalta SA, de Vos P, Zhao W, Lakey JRT. Exosome loaded immunomodulatory biomaterials alleviate local immune response in immunocompetent diabetic mice post islet xenotransplantation. Commun Biol 2021; 4:685. [PMID: 34083739 PMCID: PMC8175379 DOI: 10.1038/s42003-021-02229-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
Foreign body response (FBR) to biomaterials compromises the function of implants and leads to medical complications. Here, we report a hybrid alginate microcapsule (AlgXO) that attenuated the immune response after implantation, through releasing exosomes derived from human Umbilical Cord Mesenchymal Stem Cells (XOs). Upon release, XOs suppress the local immune microenvironment, where xenotransplantation of rat islets encapsulated in AlgXO led to >170 days euglycemia in immunocompetent mouse model of Type 1 Diabetes. In vitro analyses revealed that XOs suppressed the proliferation of CD3/CD28 activated splenocytes and CD3+ T cells. Comparing suppressive potency of XOs in purified CD3+ T cells versus splenocytes, we found XOs more profoundly suppressed T cells in the splenocytes co-culture, where a heterogenous cell population is present. XOs also suppressed CD3/CD28 activated human peripheral blood mononuclear cells (PBMCs) and reduced their cytokine secretion including IL-2, IL-6, IL-12p70, IL-22, and TNFα. We further demonstrate that XOs mechanism of action is likely mediated via myeloid cells and XOs suppress both murine and human macrophages partly by interfering with NFκB pathway. We propose that through controlled release of XOs, AlgXO provide a promising new platform that could alleviate the local immune response to implantable biomaterials.
Collapse
Affiliation(s)
- M Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California Irvine, Irvine, CA, USA
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | | | - Jennifer Cam Luong
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Shiri Li
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Rui Cao
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Hamad Alshetaiwi
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Hien Lau
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
| | - Hayk Davtyan
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - Mathew Blurton Jones
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Mahtab Jafari
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - S Armando Villalta
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, Section Immunoendocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Weian Zhao
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center; Edwards Life Sciences Center for Advanced Cardiovascular Technology; Department of Biomedical Engineering, Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Jonathan R T Lakey
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
- Department of Surgery, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
8
|
Birlutiu V, Boicean LC. Serum leptin level as a diagnostic and prognostic marker in infectious diseases and sepsis: A comprehensive literature review. Medicine (Baltimore) 2021; 100:e25720. [PMID: 33907162 PMCID: PMC8084034 DOI: 10.1097/md.0000000000025720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 02/27/2021] [Accepted: 04/11/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Infections and sepsis are common causes of morbidity and mortality, with an increasing incidence worldwide. Leptin is involved in the inflammatory process and may modulate the cytokine production, immune cell proliferation and endothelial function. There are conflicting results regarding alterations of leptin levels in infectious diseases and the outcome from sepsis.The aim of the current article is to provide an overview of the medical literature on the correlations between variations of leptin levels and infectious diseases and sepsis. METHODS We performed an extensive literature search in PubMed and Google Scholar databases, using keywords to identify articles related to leptin in infectious diseases and sepsis. Searches were referenced using medical subject headings that included "leptin," "adipokines," "sepsis," "infectious diseases," "leptin deficiency," "leptin resistance" or "hyperleptinemia." The language of publication, journal, or country were not included as limitation criteria.Articles or abstracts containing adequate information, such as age, sex, anthropometric indices, clinical presentation, comorbidities, and management were included in the study, whereas articles with insufficient clinical and demographic data were excluded. We assessed the quality of the studies selected.The final review of all databases was conducted on June 18, 2020. RESULTS We find the results from the current review to be of great importance due to the possible therapeutic role of leptin analogs in states of leptin deficiency associated with infectious diseases or sepsis.In hyperleptinemia, a therapeutic plan for obtaining leptin neutralization also needs further investigations. This could lead to the reduction of proinflammatory responses.There is a need for further studies to demonstrate the specificity and sensitivity of leptin in the early diagnosis of sepsis and the need to measure serum leptin levels in routine evaluation of the critical patient. CONCLUSION The multiple effects of leptin are of growing interest, but further studies are needed to elucidate the role of leptin signalling in infectious diseases and sepsis. Because very few human studies are reported, we recommend the need for further research.Better understanding of the pathophysiology of sepsis and the implication of circulating total leptin in this process could help physicians in managing this life-threatening condition.
Collapse
Affiliation(s)
- Victoria Birlutiu
- “Lucian Blaga” University of Sibiu, Faculty of Medicine
- Academic Emergency Hospital Sibiu—Infectious Diseases Clinic, Sibiu, Romania
| | - Loredana Camelia Boicean
- “Lucian Blaga” University of Sibiu, Faculty of Medicine
- Academic Emergency Hospital Sibiu—Infectious Diseases Clinic, Sibiu, Romania
| |
Collapse
|
9
|
Jose S, Mukherjee A, Horrigan O, Setchell KDR, Zhang W, Moreno-Fernandez ME, Andersen H, Sharma D, Haslam DB, Divanovic S, Madan R. Obeticholic acid ameliorates severity of Clostridioides difficile infection in high fat diet-induced obese mice. Mucosal Immunol 2021; 14:500-510. [PMID: 32811993 PMCID: PMC7889747 DOI: 10.1038/s41385-020-00338-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/18/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
Abstract
Severe Clostridiodes difficile infection (CDI) is life-threatening and responds poorly to treatment. Obesity is associated with development of severe CDI. Therefore, to define the mechanisms that exacerbate disease severity, we examined CDI pathogenesis in high-fat diet (HFD)-fed obese mice. Compared to control mice, HFD-fed mice failed to clear C. difficile bacteria which resulted in protracted diarrhea, weight loss and colonic damage. After infection, HFD-induced obese mice had an intestinal bile acid (BA) pool that was dominated by primary BAs which are known promoters of C. difficile spore germination, and lacked secondary BAs that inhibit C. difficile growth. Concurrently, synthesis of primary BAs from liver was significantly increased in C. difficile-infected HFD-fed mice. A key pathway that regulates hepatic BA synthesis is via feedback inhibition from intestinal Farnesoid X receptors (FXRs). Our data reveal that the proportion of FXR agonist BAs to FXR antagonist BAs in the intestinal lumen was significantly reduced in HFD-fed mice after CDI. Treatment of HFD-fed mice with an FXR agonist Obeticholic acid, resulted in decreased primary BA synthesis, fewer C. difficile bacteria and better CDI outcomes. Thus, OCA treatment holds promise as a therapy for severe CDI.
Collapse
Affiliation(s)
- Shinsmon Jose
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Anindita Mukherjee
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Olivia Horrigan
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Kenneth D R Setchell
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - Wujuan Zhang
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Heidi Andersen
- Department of Pediatric Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Divya Sharma
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
| | - David B Haslam
- Department of Pediatric Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Rajat Madan
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45220, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA.
| |
Collapse
|
10
|
Kiernan K, MacIver NJ. The Role of the Adipokine Leptin in Immune Cell Function in Health and Disease. Front Immunol 2021; 11:622468. [PMID: 33584724 PMCID: PMC7878386 DOI: 10.3389/fimmu.2020.622468] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022] Open
Abstract
Leptin is a critical mediator of the immune response to changes in overall nutrition. Leptin is produced by adipocytes in proportion to adipose tissue mass and is therefore increased in obesity. Despite having a well-described role in regulating systemic metabolism and appetite, leptin displays pleiotropic actions, and it is now clear that leptin has a key role in influencing immune cell function. Indeed, many immune cells have been shown to respond to leptin directly via the leptin receptor, resulting in a largely pro-inflammatory phenotype. Understanding the role of adipose-tissue derived mediators in inflammation is critical to determining the pathophysiology of multiple obesity-associated diseases, such as type 2 diabetes, autoimmune disease, and infection. This review, therefore, focuses on the latest data regarding the role of leptin in modulating inflammation.
Collapse
Affiliation(s)
- Kaitlin Kiernan
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Nancie J. MacIver
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
11
|
A centric view of JAK/STAT5 in intestinal homeostasis, infection, and inflammation. Cytokine 2021; 139:155392. [PMID: 33482575 PMCID: PMC8276772 DOI: 10.1016/j.cyto.2020.155392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Cytokines, growth factors or hormones take action through the JAK/STAT5 signaling pathway, which plays a critical role in regulating the intestinal response to infection and inflammation. However, the way in which STAT5 regulates intestinal epithelial compartment is largely ignored due to the lack of genetic tools for proper exploration and because the two STAT5 transcription factors (STAT5A and STAT5B) have some redundant but also distinct functions. In this review article, by focusing on STAT5 functions in the intestinal undifferentiated and differentiated epithelia, we discuss major advances of the growth factor/cytokine-JAK/STAT5 research in view of intestinal mucosal inflammation and immunity. We highlight the gap in the research of the intestinal STAT5 signaling to anticipate the gastrointestinal explorative insights. Furthermore, we address the critical questions to illuminate how STAT5 signaling influences intestinal epithelial cell differentiation and stem cell regeneration during homeostasis and injury. Overall, our article provides a centric view of the relevance of the relationship between chronic inflammatory diseases and JAK/STAT5 pathway and it also gives an example of how chronic infection and inflammation pirate STAT5 signaling to worsen intestinal injuries. Importantly, our review suggests how to protect a wound healing from gastrointestinal diseases by modulating intestinal STAT5.
Collapse
|
12
|
Chang ML, Yang Z, Yang SS. Roles of Adipokines in Digestive Diseases: Markers of Inflammation, Metabolic Alteration and Disease Progression. Int J Mol Sci 2020; 21:E8308. [PMID: 33167521 PMCID: PMC7663948 DOI: 10.3390/ijms21218308] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is a highly dynamic endocrine tissue and constitutes a central node in the interorgan crosstalk network through adipokines, which cause pleiotropic effects, including the modulation of angiogenesis, metabolism, and inflammation. Specifically, digestive cancers grow anatomically near adipose tissue. During their interaction with cancer cells, adipocytes are reprogrammed into cancer-associated adipocytes and secrete adipokines to affect tumor cells. Moreover, the liver is the central metabolic hub. Adipose tissue and the liver cooperatively regulate whole-body energy homeostasis via adipokines. Obesity, the excessive accumulation of adipose tissue due to hyperplasia and hypertrophy, is currently considered a global epidemic and is related to low-grade systemic inflammation characterized by altered adipokine regulation. Obesity-related digestive diseases, including gastroesophageal reflux disease, Barrett's esophagus, esophageal cancer, colon polyps and cancer, non-alcoholic fatty liver disease, viral hepatitis-related diseases, cholelithiasis, gallbladder cancer, cholangiocarcinoma, pancreatic cancer, and diabetes, might cause specific alterations in adipokine profiles. These patterns and associated bases potentially contribute to the identification of prognostic biomarkers and therapeutic approaches for the associated digestive diseases. This review highlights important findings about altered adipokine profiles relevant to digestive diseases, including hepatic, pancreatic, gastrointestinal, and biliary tract diseases, with a perspective on clinical implications and mechanistic explorations.
Collapse
Affiliation(s)
- Ming-Ling Chang
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
- Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Zinger Yang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA;
| | - Sien-Sing Yang
- Liver Center, Cathay General Hospital Medical Center, Taipei 10630, Taiwan;
| |
Collapse
|
13
|
Matsumura S, Kurashima Y, Murasaki S, Morimoto M, Arai F, Saito Y, Katayama N, Kim D, Inagaki Y, Kudo T, Ernst PB, Shimizu T, Kiyono H. Stratified layer analysis reveals intrinsic leptin stimulates cryptal mesenchymal cells for controlling mucosal inflammation. Sci Rep 2020; 10:18351. [PMID: 33110098 PMCID: PMC7591933 DOI: 10.1038/s41598-020-75186-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/07/2020] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal cells in the crypt play indispensable roles in the maintenance of intestinal epithelial homeostasis through their contribution to the preservation of stem cells. However, the acquisition properties of the production of stem cell niche factors by the mesenchymal cells have not been well elucidated, due to technical limitations regarding the isolation and subsequent molecular and cellular analyses of cryptal mesenchymal cells. To evaluate the function of mesenchymal cells located at the large intestinal crypt, we established a novel method through which cells are harvested according to the histologic layers of mouse colon, and we compared cellular properties between microenvironmental niches, the luminal mucosa and crypts. The gene expression pattern in the cryptal mesenchymal cells showed that receptors of the hormone/cytokine leptin were highly expressed, and we found a decrease in Wnt2b expression under conditions of leptin receptor deficiency, which also induced a delay in cryptal epithelial proliferation. Our novel stratified layer isolation strategies thus revealed new microenvironmental characteristics of colonic mesenchymal cells, including the intrinsic involvement of leptin in the control of mucosal homeostasis.
Collapse
Affiliation(s)
- Seiichi Matsumura
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan.,Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Pediatrics, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan. .,Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,Division of Gastroenterology, Department of Medicine, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA, 92093-0956, USA. .,Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, San Diego, CA, 92093-0956, USA.
| | - Sayuri Murasaki
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Masako Morimoto
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Fujimi Arai
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yukari Saito
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8670, Japan
| | - Nana Katayama
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Dayoung Kim
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, Japan
| | - Takahiro Kudo
- Department of Pediatrics, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Peter B Ernst
- Division of Gastroenterology, Department of Medicine, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA, 92093-0956, USA.,Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, San Diego, CA, 92093-0956, USA.,Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA, 92093-0956, USA
| | - Toshiaki Shimizu
- Department of Pediatrics, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroshi Kiyono
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Division of Gastroenterology, Department of Medicine, CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California, San Diego, CA, 92093-0956, USA
| |
Collapse
|
14
|
Xu Q, Li Y, Zheng Y, Chen Y, Xu X, Wang M. Clostridium difficile toxin B-induced colonic inflammation is mediated by the FOXO3/PPM1B pathway in fetal human colon epithelial cells. Am J Transl Res 2020; 12:6204-6219. [PMID: 33194024 PMCID: PMC7653611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Clostridium difficile (C. difficile) toxin B (TcdB) is as an inflammatory enterotoxin that accounts for manifestations of widespread healthcare-associated C. difficile infection, including colonic inflammation. The present work explored the molecular mechanism by which TcdB activates innate immunity and stimulates pro-inflammatory cytokine release. Fetal human colon epithelial cells (FHCs) were treated with recombinant TcdB protein. Cell growth inhibition and apoptosis were measured with Cell Counting Kit-8 and Annexin V-fluorescein isothiocyanate Apoptosis Detection Kit, respectively. Flow cytometry analysis was also performed. Inflammatory cytokine induction was determined with enzykeme-linked immunosorbent assay analyses. Protein expression was assessed by western blot analysis. Gene overexpression and knockdown were performed with lentiviral transduction. Real-time quantitative polymerase chain reaction was used to examine gene expression. Dual-luciferase reporter assays and chromatin immunoprecipitation were implemented to explore transcriptional regulation. Mouse colon tissues were analyzed with hematoxylin and eosin staining. The results show that TcdB-induced cell growth and apoptosis and enhanced expression of interleukin-6 and tumor necrosis factor alpha in FHCs. We identified protein phosphatase magnesium-dependent 1B (PPM1B) as the key mediator promoting the phosphorylation of nuclear factor-κB p65, which accounted for the increase in pro-inflammatory cytokines. The findings demonstrate that PPM1B expression is directly regulated by the AKT/FOXO3 signaling pathway in FHCs. We confirmed the molecular mechanism with in vivo studies using a mouse model infected with C. difficile and treated with a phosphoinositide 3-kinase/AKT signaling inhibitor. In conclusion, TcdB induces inflammation in human colon epithelial cells by regulating the AKT/FOXO3/PPM1B pathway.
Collapse
Affiliation(s)
- Qingqing Xu
- Institute of Antibiotics, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health and Family Planning CommisionShanghai 200040, China
| | - Ying Li
- Institute of Antibiotics, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health and Family Planning CommisionShanghai 200040, China
| | - Yuejuan Zheng
- Department of Immunology and Microbiology, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Yijian Chen
- Institute of Antibiotics, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health and Family Planning CommisionShanghai 200040, China
| | - Xiaogang Xu
- Institute of Antibiotics, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health and Family Planning CommisionShanghai 200040, China
| | - Minggui Wang
- Institute of Antibiotics, Huashan Hospital, Fudan UniversityShanghai 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health and Family Planning CommisionShanghai 200040, China
| |
Collapse
|
15
|
Pérez-Pérez A, Sánchez-Jiménez F, Vilariño-García T, Sánchez-Margalet V. Role of Leptin in Inflammation and Vice Versa. Int J Mol Sci 2020; 21:E5887. [PMID: 32824322 PMCID: PMC7460646 DOI: 10.3390/ijms21165887] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammation is an essential immune response for the maintenance of tissue homeostasis. In a general sense, acute and chronic inflammation are different types of adaptive response that are called into action when other homeostatic mechanisms are insufficient. Although considerable progress has been made in understanding the cellular and molecular events that are involved in the acute inflammatory response to infection and tissue injury, the causes and mechanisms of systemic chronic inflammation are much less known. The pathogenic capacity of this type of inflammation is puzzling and represents a common link of the multifactorial diseases, such as cardiovascular diseases and type 2 diabetes. In recent years, interest has been raised by the discovery of novel mediators of inflammation, such as microRNAs and adipokines, with different effects on target tissues. In the present review, we discuss the data emerged from research of leptin in obesity as an inflammatory mediator sustaining multifactorial diseases and how this knowledge could be instrumental in the design of leptin-based manipulation strategies to help restoration of abnormal immune responses. On the other direction, chronic inflammation, either from autoimmune or infectious diseases, or impaired microbiota (dysbiosis) may impair the leptin response inducing resistance to the weight control, and therefore it may be a cause of obesity. Thus, we are reviewing the published data regarding the role of leptin in inflammation, and the other way around, the role of inflammation on the development of leptin resistance and obesity.
Collapse
Affiliation(s)
- Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain; (F.S.-J.); (T.V.-G.)
| | | | | | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain; (F.S.-J.); (T.V.-G.)
| |
Collapse
|
16
|
Bekeredjian-Ding I. Challenges for Clinical Development of Vaccines for Prevention of Hospital-Acquired Bacterial Infections. Front Immunol 2020; 11:1755. [PMID: 32849627 PMCID: PMC7419648 DOI: 10.3389/fimmu.2020.01755] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
Increasing antibiotic resistance in bacteria causing endogenous infections has entailed a need for innovative approaches to therapy and prophylaxis of these infections and raised a new interest in vaccines for prevention of colonization and infection by typically antibiotic resistant pathogens. Nevertheless, there has been a long history of failures in late stage clinical development of this type of vaccines, which remains not fully understood. This article provides an overview on present and past vaccine developments targeting nosocomial bacterial pathogens; it further highlights the specific challenges associated with demonstrating clinical efficacy of these vaccines and the facts to be considered in future study designs. Notably, these vaccines are mainly applied to subjects with preexistent immunity to the target pathogen, transient or chronic immunosuppression and ill-defined microbiome status. Unpredictable attack rates and changing epidemiology as well as highly variable genetic and immunological strain characteristics complicate the development. In views of the clinical need, re-thinking of the study designs and expectations seems warranted: first of all, vaccine development needs to be footed on a clear rationale for choosing the immunological mechanism of action and the optimal time point for vaccination, e.g., (1) prevention (or reduction) of colonization vs. prevention of infection and (2) boosting of a preexistent immune response vs. altering the quality of the immune response. Furthermore, there are different, probably redundant, immunological and microbiological defense mechanisms that provide protection from infection. Their interplay is not well-understood but as a consequence their effect might superimpose vaccine-mediated resolution of infection and lead to failure to demonstrate efficacy. This implies that improved characterization of patient subpopulations within the trial population should be obtained by pro- and retrospective analyses of trial data on subject level. Statistical and systems biology approaches could help to define immune and microbiological biomarkers that discern populations that benefit from vaccination from those where vaccines might not be effective.
Collapse
Affiliation(s)
- Isabelle Bekeredjian-Ding
- Division of Microbiology, Langen, Germany.,Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
17
|
Ok M, Yildiz R, Hatipoglu F, Baspinar N, Ider M, Üney K, Ertürk A, Durgut MK, Terzi F. Use of intestine-related biomarkers for detecting intestinal epithelial damage in neonatal calves with diarrhea. Am J Vet Res 2020; 81:139-146. [PMID: 31985285 DOI: 10.2460/ajvr.81.2.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the usefulness of intestinal biomarkers in determining the presence of intestinal epithelial damage in neonatal calves with diarrhea caused by 4 etiologic agents. ANIMALS 40 neonatal calves that were healthy (n = 10) or had diarrhea (30). PROCEDURES The study was a cross-sectional study. Results of hematologic analyses and serum concentrations of intestinal fatty acid-binding protein (I-FABP), liver fatty acid-binding protein (L-FABP), trefoil factor 3 (TFF-3), Claudin-3 (CLDN-3), γ-enteric smooth muscle actin (ACTG2), intestinal alkaline phosphatase (IAP), interleukin-8 (IL-8), platelet-activating factor (PAF), and leptin (LP) were compared among calves grouped according to whether they were healthy (control group; G-1) or had diarrhea caused by K99 Escherichia coli (G-2; n = 10), bovine rota- or coronavirus (G-3; 5 each), or Cryptosporidium spp (G-4; 10). RESULTS Across the 3 time points at which blood samples were obtained and evaluated, the groups of calves with diarrhea generally had markedly higher mean serum concentrations of L-FABP, TFF-3, IAP, IL-8, and LP, compared with the control group. In addition, G-2 also consistently had markedly higher mean serum concentrations of I-FAB and ACTG2 and lower mean serum concentrations of CLDN-3, compared with the control group. CONCLUSIONS AND CLINICAL RELEVANCE Results indicated that degree of intestinal epithelial damage differed among calves grouped by the etiologic agent of diarrhea and that such damage might have been more severe in calves with diarrhea caused by K99 E coli. Additionally, our results indicated that serum concentrations of I-FABP, L-FABP, TFF-3, IAP, IL-8, ACTG2, LP, and CLDN-3 were useful biomarkers of intestinal epithelial damage in calves of the present study.
Collapse
|
18
|
Abstract
Clostridioides difficile infection is the most common health care-associated infection in the United States with more than 20% patients experiencing symptomatic recurrence. The complex nature of host-bacterium interactions makes it difficult to predict the course of the disease based solely on clinical parameters. In the present study, we built a robust prediction model using representative plasma biomarkers and clinical parameters for 90-day all-cause mortality. Risk prediction based on immune biomarkers and clinical variables may contribute to treatment selection for patients as well as provide insight into the role of immune system in C. difficile pathogenesis. There is a pressing need for biomarker-based models to predict mortality from and recurrence of Clostridioides difficile infection (CDI). Risk stratification would enable targeted interventions such as fecal microbiota transplant, antitoxin antibodies, and colectomy for those at highest risk. Because severity of CDI is associated with the immune response, we immune profiled patients at the time of diagnosis. The levels of 17 cytokines in plasma were measured in 341 CDI inpatients. The primary outcome of interest was 90-day mortality. Increased tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), C-C motif chemokine ligand 5 (CCL-5), suppression of tumorigenicity 2 receptor (sST-2), IL-8, and IL-15 predicted mortality by univariate analysis. After adjusting for demographics and clinical characteristics, the mortality risk (as indicated by the hazard ratio [HR]) was higher for patients in the top 25th percentile for TNF-α (HR = 8.35, P = 0.005) and IL-8 (HR = 4.45, P = 0.01) and lower for CCL-5 (HR = 0.18, P ≤ 0.008). A logistic regression risk prediction model was developed and had an area under the receiver operating characteristic curve (AUC) of 0.91 for 90-day mortality and 0.77 for 90-day recurrence. While limited by being single site and retrospective, our work resulted in a model with a substantially greater predictive ability than white blood cell count. In conclusion, immune profiling demonstrated differences between patients in their response to CDI, offering the promise for precision medicine individualized treatment.
Collapse
|
19
|
Exogenous leptin reinforces intestinal barrier function and protects from colitis. Pharmacol Res 2019; 147:104356. [DOI: 10.1016/j.phrs.2019.104356] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/19/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
|
20
|
Roberts SA, Brabin L, Diallo S, Gies S, Nelson A, Stewart C, Swinkels DW, Geurts-Moespot AJ, Kazienga A, Ouedraogo S, D'Alessandro U, Tinto H, Brabin BJ. Mucosal lactoferrin response to genital tract infections is associated with iron and nutritional biomarkers in young Burkinabé women. Eur J Clin Nutr 2019; 73:1464-1472. [PMID: 31168085 PMCID: PMC6842079 DOI: 10.1038/s41430-019-0444-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/29/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND/OBJECTIVES The iron-binding affinity of vaginal lactoferrin (Lf) reduces iron available to genital pathogens. We describe host reproductive, nutritional, infection and iron biomarker profiles affecting vaginal Lf concentration in young nulliparous and primigravid women in Burkina Faso. SUBJECTS/METHODS Vaginal eluates from women who had participated in a randomized, controlled periconceptional iron supplementation trial were used to measure Lf using a competitive double-sandwich ELISA. For this analysis samples from both trial arms were combined and pregnant and non-pregnant cohorts compared. Following randomization Lf was measured after 18 months (end assessment) for women remaining non-pregnant, and at two antenatal visits for those becoming pregnant. Associations between log Lf levels and demographic, anthropometric, infection and iron biomarker variables were assessed using linear mixed models. RESULTS Lf samples were available for 712 non-pregnant women at end assessment and for 303 women seen at an antenatal visit. Lf concentrations of pregnant women were comparable to those of non-pregnant, sexually active women. Lf concentration increased with mid-upper-arm circumference, (P = 0.047), body mass index (P = 0.018), Trichomonas vaginalis (P < 0.001) infection, bacterial vaginosis (P < 0.001), serum C-reactive protein (P = 0.048) and microbiota community state types III/IV. Adjusted Lf concentration was positively associated with serum hepcidin (P = 0.047), serum ferritin (P = 0.018) and total body iron stores (P = 0.042). There was evidence that some women maintained persistently high or low Lf concentrations from before, and through, pregnancy. CONCLUSION Lf concentrations increased with genital infection, higher BMI, MUAC, body iron stores and hepcidin, suggesting nutritional and iron status influence homeostatic mechanisms controlling vaginal Lf responses.
Collapse
Affiliation(s)
- S A Roberts
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary Care, Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - L Brabin
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - S Diallo
- Clinical Research Unit of Nanoro (URCN/IRSS), Nanoro, Burkina Faso
| | - S Gies
- Department of Biomedical Sciences, Prince Leopold Institute of Tropical Medicine, Antwerp, Belgium and Medical Mission Institute, Würzburg, Germany
| | - A Nelson
- Faculty of Health and Life Sciences, University of Northumbria, Northumberland Building, Newcastle-upon-Tyne, UK
| | - C Stewart
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - D W Swinkels
- Department of Laboratory Medicine (TLM 830), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands.,Hepcidinanalysis.com., Nijmegen, The Netherlands
| | - A J Geurts-Moespot
- Department of Laboratory Medicine (TLM 830), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - A Kazienga
- Clinical Research Unit of Nanoro (URCN/IRSS), Nanoro, Burkina Faso
| | - S Ouedraogo
- Clinical Research Unit of Nanoro (URCN/IRSS), Nanoro, Burkina Faso
| | - U D'Alessandro
- Medical Research Council Unit (MRC), Fajara, The Gambia.,London School of Hygiene and Tropical Medicine, London, UK
| | - H Tinto
- Clinical Research Unit of Nanoro (URCN/IRSS), Nanoro, Burkina Faso
| | - B J Brabin
- Liverpool School of Tropical Medicine, and Institute of Infection and Global Health, University of Liverpool, Liverpool, UK. .,Global Child Health Group, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Cortese L, Terrazzano G, Pelagalli A. Leptin and Immunological Profile in Obesity and Its Associated Diseases in Dogs. Int J Mol Sci 2019; 20:2392. [PMID: 31091785 PMCID: PMC6566566 DOI: 10.3390/ijms20102392] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/29/2022] Open
Abstract
Growing scientific evidence has unveiled increased incidences of obesity in domestic animals and its influence on a plethora of associated disorders. Leptin, an adipokine regulating body fat mass, represents a key molecule in obesity, able to modulate immune responses and foster chronic inflammatory response in peripheral tissues. High levels of cytokines and inflammatory markers suggest an association between inflammatory state and obesity in dogs, highlighting the parallelism with humans. Canine obesity is a relevant disease always accompanied with several health conditions such as inflammation, immune-dysregulation, insulin resistance, pancreatitis, orthopaedic disorders, cardiovascular disease, and neoplasia. However, leptin involvement in many disease processes in veterinary medicine is poorly understood. Moreover, hyperleptinemia as well as leptin resistance occur with cardiac dysfunction as a consequence of altered cardiac mitochondrial metabolism in obese dogs. Similarly, leptin dysregulation seems to be involved in the pancreatitis pathophysiology. This review aims to examine literature concerning leptin and immunological status in obese dogs, in particular for the aspects related to obesity-associated diseases.
Collapse
Affiliation(s)
- Laura Cortese
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy.
| | - Giuseppe Terrazzano
- Department of Science, University of Basilicata, 85100 Potenza, Italy.
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Naples, Italy.
- Institute of Biostructures and Bioimages (IBB), National Research Council (CNR), 80131 Naples, Italy.
| |
Collapse
|
22
|
Mota-Zamorano S, Luna E, Garcia-Pino G, González LM, Gervasini G. Variability in the leptin receptor gene and other risk factors for post-transplant diabetes mellitus in renal transplant recipients. Ann Med 2019; 51:164-173. [PMID: 31046466 PMCID: PMC7857488 DOI: 10.1080/07853890.2019.1614656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Post-transplant diabetes mellitus (PTDM) is one of the main complications after kidney transplantation. It is known that leptin plays an important role in glucose metabolism and mutations in the leptin receptor gene (LEPR) are responsible for different complications in renal transplant recipients. We aimed to analyse the association of polymorphisms in LEPR with the development of PTDM in these patients. Methods: A total of 315 renal transplant recipients were genotyped for the Lys109Arg, Gln223Arg and Lys656Asn polymorphisms. The impact of these genetic variables together with other clinical and demographic parameters on PTDM risk was evaluated in a multivariate regression analysis. Results: The 223Arg variant showed a significant association with PTDM risk [OR = 3.26 (1.35-7.85), p = 0.009] after correcting for multiple testing. Carriers of this variant also showed higher BMI values (26.95 ± 4.23) than non-carriers (25.67 ± 4.43, p = 0.025). In addition, it was BMI at transplant and not the BMI increment in the first year after grafting that was associated with PTDM (p > 0.00001). Haplotype analyses did not reveal significant associations. Conclusions: Our result show, for the first time to our knowledge, that genetic variability in the LEPR may contribute significantly to the risk for PTDM in renal transplant recipients. KEY MESSAGES The LEPR Gln223Arg polymorphism significantly contributes to the development of PTDM in renal transplant recipients. The effect of the 223Arg variant on PTDM is strongly modulated by the age of the recipient. The 223Arg variant in the leptin receptor is related to higher BMI in renal transplant recipients.
Collapse
Affiliation(s)
- Sonia Mota-Zamorano
- a Department of Medical and Surgical Therapeutics, Division of Pharmacology, Medical School , University of Extremadura , Badajoz , Spain
| | - Enrique Luna
- b Service of Nephrology , Badajoz University Hospital , Badajoz , Spain
| | | | - Luz M González
- a Department of Medical and Surgical Therapeutics, Division of Pharmacology, Medical School , University of Extremadura , Badajoz , Spain
| | - Guillermo Gervasini
- a Department of Medical and Surgical Therapeutics, Division of Pharmacology, Medical School , University of Extremadura , Badajoz , Spain
| |
Collapse
|
23
|
Molecular dynamic (MD) studies on Gln233Arg (rs1137101) polymorphism of leptin receptor gene and associated variations in the anthropometric and metabolic profiles of Saudi women. PLoS One 2019; 14:e0211381. [PMID: 30763324 PMCID: PMC6375553 DOI: 10.1371/journal.pone.0211381] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 01/12/2019] [Indexed: 01/07/2023] Open
Abstract
The Gln233Arg (A>G; rs1137101) polymorphism of the leptin receptor gene (LEPR) has been investigated extensively and is reported to be associated with different metabolic states. In this investigation, we aimed to study the frequency of Gln233Arg genotypes and alleles in a group of Saudi women stratified by their body mass index (BMI), to correlate the LEPR genotypes with variations in anthropometric, lipid and hormonal parameters and to investigate conformational and structural variations in the mutant LEPR using molecular dynamic (MD) investigations. The study group included 122 Saudi women (normal weight = 60; obese = 62) attending the clinics for a routine checkup. Anthropometric data: height, weight, waist and hip circumference were recorded and fasting serum sample was used to estimate glucose, lipids, ghrelin, leptin and insulin. BMI, W/H ratio, and HOMA-IR values were calculated. Whole blood sample was used to extract DNA; exon 6 of the LEPR gene was amplified by PCR and sequencing was conducted on an ABI 3100 Avant Genetic Analyser. Molecular Dynamic Simulation studies were carried out using different softwares. The results showed the presence of all three genotypes of Gln233Arg in Saudi women, but the frequencies were significantly different when compared to reports from some populations. No differences were seen in the genotype and allele frequencies between the normal weight and obese women. Stratification by the genotypes showed significantly higher BMI, waist and hip circumference, leptin, insulin, fasting glucose and HOMA-IR and lower ghrelin levels in obese women carrying the GG genotype. Even in the normal weight group, individuals with GG genotype had higher BMI, waist and hip circumference and significantly lower ghrelin levels. The MD studies showed a significant effect of the Gln/Arg substitution on the conformation, flexibility, root-mean-square fluctuation (RMSF), radius of gyration (Rg) values, solvent-accessible surface area (SASA) and number of inter- and intra-molecular H-bonds. The results suggest that the structural changes brought about by the mutation, influence the signaling pathways by some unknown mechanism, which may be contributing to the abnormalities seen in the individuals carrying the G allele of rs1137101.
Collapse
|
24
|
Maurya R, Bhattacharya P, Dey R, Nakhasi HL. Leptin Functions in Infectious Diseases. Front Immunol 2018; 9:2741. [PMID: 30534129 PMCID: PMC6275238 DOI: 10.3389/fimmu.2018.02741] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 11/07/2018] [Indexed: 01/26/2023] Open
Abstract
Leptin, a pleiotropic protein has long been recognized to play an important role in the regulation of energy homeostasis, metabolism, neuroendocrine function, and other physiological functions through its effects on the central nervous system (CNS) and peripheral tissues. Leptin is secreted by adipose tissue and encoded by the obese (ob) gene. Leptin acts as a central mediator which regulates immunity as well as nutrition. Importantly, leptin can modulate both innate and adaptive immune responses. Leptin deficiency/resistance is associated with dysregulation of cytokine production, increased susceptibility toward infectious diseases, autoimmune disorders, malnutrition and inflammatory responses. Malnutrition induces a state of immunodeficiency and an inclination to death from communicable diseases. Infectious diseases are the disease of poor who invariably suffer from malnutrition that could result from reduced serum leptin levels. Thus, leptin has been placed at the center of many interrelated functions in various pathogenic conditions, such as bacterial, viruses and parasitic infections. We review herein, the recent advances on the role of leptin in malnutrition in pathogenesis of infectious diseases with a particular emphasis on parasitic diseases such as Leishmaniasis, Trypanosomiasis, Amoebiasis, and Malaria.
Collapse
Affiliation(s)
- Radheshyam Maurya
- Department of Animal Biology, School of Life Science, University of Hyderabad, Hyderabad, India
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
25
|
Jose S, Mukherjee A, Abhyankar MM, Leng L, Bucala R, Sharma D, Madan R. Neutralization of macrophage migration inhibitory factor improves host survival after Clostridium difficile infection. Anaerobe 2018; 53:56-63. [PMID: 29944928 PMCID: PMC6309669 DOI: 10.1016/j.anaerobe.2018.06.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/16/2022]
Abstract
Clostridium difficile is an important cause of nosocomial diarrhea in the western world. Toxins (A, B, and binary toxins) generated by C. difficile bacteria damage intestinal epithelial cells. Hallmarks of host response to C. difficile infection (CDI) include upregulation of inflammatory mediators and tissue infiltration by immune cells. Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine that is known to enhance the host immune response to infectious pathogens. Additionally, MIF can adversely impact host survival to numerous infections. The role of MIF in the pathogenesis of CDI remains poorly understood. Here, we show that patients with CDI had significantly higher circulating MIF compared to patients who had diarrhea but tested negative for C. difficile (non-CDI controls). Similarly, in a mouse model, C. difficile challenge significantly increased levels of plasma and tissue MIF. Antibody-mediated depletion of MIF decreased C. difficile-induced inflammatory responses, clinical disease, and mortality. Together, these results uncover a potential role for MIF in exacerbating CDI and suggest that use of anti-MIF antibodies may represent a therapeutic strategy to curb host inflammatory responses and improve disease outcomes in CDI.
Collapse
Affiliation(s)
- Shinsmon Jose
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Anindita Mukherjee
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Mayuresh M Abhyankar
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Lin Leng
- Departments of Internal Medicine, Yale University, New Haven, CT, USA
| | - Richard Bucala
- Departments of Internal Medicine, Yale University, New Haven, CT, USA
| | - Divya Sharma
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Rajat Madan
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA; Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
26
|
Alti D, Sambamurthy C, Kalangi SK. Emergence of Leptin in Infection and Immunity: Scope and Challenges in Vaccines Formulation. Front Cell Infect Microbiol 2018; 8:147. [PMID: 29868503 PMCID: PMC5954041 DOI: 10.3389/fcimb.2018.00147] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/20/2018] [Indexed: 01/01/2023] Open
Abstract
Deficiency of leptin (ob/ob) and/or desensitization of leptin signaling (db/db) and elevated expression of suppressor of cytokine signaling-3 (SOCS3) reported in obesity are also reported in a variety of pathologies including hypertriglyceridemia, insulin resistance, and malnutrition as the risk factors in host defense system. Viral infections cause the elevated SOCS3 expression, which inhibits leptin signaling. It results in immunosuppression by T-regulatory cells (Tregs). The host immunity becomes incompetent to manage pathogens' attack and invasion, which results in the accelerated infections and diminished vaccine-specific antibody response. Leptin was successfully used as mucosal vaccine adjuvant against Rhodococcus equi. Leptin induced the antibody response to Helicobacter pylori vaccination in mice. An integral leptin signaling in mucosal gut epithelial cells offered resistance against Clostridium difficile and Entameoba histolytica infections. We present in this review, the intervention of leptin in lethal diseases caused by microbial infections and propose the possible scope and challenges of leptin as an adjuvant tool in the development of effective vaccines.
Collapse
Affiliation(s)
- Dayakar Alti
- School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | | - Suresh K Kalangi
- School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
27
|
Jose S, Abhyankar MM, Mukherjee A, Xue J, Andersen H, Haslam DB, Madan R. Leptin receptor q223r polymorphism influences neutrophil mobilization after Clostridium difficile infection. Mucosal Immunol 2018; 11:947-957. [PMID: 29363668 PMCID: PMC5976520 DOI: 10.1038/mi.2017.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/11/2017] [Indexed: 02/04/2023]
Abstract
Clostridium difficile is the leading cause of nosocomial infections in the United States. Clinical disease outcomes after C. difficile infection (CDI) are dependent on intensity of host inflammatory responses. Specifically, peak peripheral white blood cell (WBC) count >20 × 109 l-1 is an indicator of adverse outcomes in CDI patients, and is associated with higher 30-day mortality. We show that homozygosity for a common single nucleotide polymorphism (Q to R mutation in leptin receptor that is present in up to 50% of people), significantly increases the risk of having peak peripheral WBC count >20 × 109 l-1 (odds ratio=5.41; P=0.0023) in CDI patients. In a murine model of CDI, we demonstrate that mice homozygous for the same single nucleotide polymorphism (RR mice) have more blood and tissue leukocytes (specifically neutrophils), exaggerated tissue inflammation, and higher mortality as compared with control mice, despite similar pathogen burden. Further, we show that neutrophilia in RR mice is mediated by gut microbiota-directed expression of CXC chemokine receptor 2 (CXCR2), which promotes the release of neutrophils from bone marrow reservoir. Overall these studies provide novel mechanistic insights into the role of human genetic polymorphisms and gut microbiota in regulating the fundamental biological process of CDI-induced neutrophilia.
Collapse
Affiliation(s)
- Shinsmon Jose
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA
| | - Mayuresh M. Abhyankar
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Anindita Mukherjee
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA
| | - Jianli Xue
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA
| | - Heidi Andersen
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - David B. Haslam
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rajat Madan
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA,Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45267, USA,Correspondence:
| |
Collapse
|
28
|
Thaiss CA, Levy M, Grosheva I, Zheng D, Soffer E, Blacher E, Braverman S, Tengeler AC, Barak O, Elazar M, Ben-Zeev R, Lehavi-Regev D, Katz MN, Pevsner-Fischer M, Gertler A, Halpern Z, Harmelin A, Aamar S, Serradas P, Grosfeld A, Shapiro H, Geiger B, Elinav E. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 2018. [PMID: 29519916 DOI: 10.1126/science.aar3318] [Citation(s) in RCA: 601] [Impact Index Per Article: 85.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity, diabetes, and related manifestations are associated with an enhanced, but poorly understood, risk for mucosal infection and systemic inflammation. Here, we show in mouse models of obesity and diabetes that hyperglycemia drives intestinal barrier permeability, through GLUT2-dependent transcriptional reprogramming of intestinal epithelial cells and alteration of tight and adherence junction integrity. Consequently, hyperglycemia-mediated barrier disruption leads to systemic influx of microbial products and enhanced dissemination of enteric infection. Treatment of hyperglycemia, intestinal epithelial-specific GLUT2 deletion, or inhibition of glucose metabolism restores barrier function and bacterial containment. In humans, systemic influx of intestinal microbiome products correlates with individualized glycemic control, indicated by glycated hemoglobin levels. Together, our results mechanistically link hyperglycemia and intestinal barrier function with systemic infectious and inflammatory consequences of obesity and diabetes.
Collapse
Affiliation(s)
- Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maayan Levy
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Inna Grosheva
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Danping Zheng
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eliran Soffer
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Blacher
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sofia Braverman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Anouk C Tengeler
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Barak
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.,Department of Obstetrics and Gynecology, Kaplan Medical Center, Rehovot, affiliated with the Hebrew University and Hadassah School of Medicine, Jerusalem, Israel
| | - Maya Elazar
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Rotem Ben-Zeev
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dana Lehavi-Regev
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Meirav N Katz
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Arieh Gertler
- The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, Israel
| | - Zamir Halpern
- Sackler Faculty of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Digestive Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Suhail Aamar
- Department of Medicine, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Patricia Serradas
- INSERM Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Cités, UPD Univ. Paris 05, CNRS, IHU ICAN, Paris, France
| | - Alexandra Grosfeld
- INSERM Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Cités, UPD Univ. Paris 05, CNRS, IHU ICAN, Paris, France
| | - Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
29
|
Wang L, Cao J, Li C, Zhang L. IL-27/IL-27 Receptor Signaling Provides Protection in Clostridium difficile-Induced Colitis. J Infect Dis 2017; 217:198-207. [DOI: 10.1093/infdis/jix581] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 11/04/2017] [Indexed: 01/03/2023] Open
|
30
|
Leptin regulates Granzyme-A, PD-1 and CTLA-4 expression in T cell to control visceral leishmaniasis in BALB/c Mice. Sci Rep 2017; 7:14664. [PMID: 29116252 PMCID: PMC5676676 DOI: 10.1038/s41598-017-15288-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/24/2017] [Indexed: 01/01/2023] Open
Abstract
Visceral leishmaniasis (VL) is responsible for several deaths in malnourished children accompanied by diminished circulating leptin and impaired cell-mediated immunity. Typically, leptin deficiency is associated with the Th2 polarization that markedly coincides with the pathogenesis of VL. The aim of the present study was to unravel the prophylactic role of leptin in malnutrition-coupled VL mice. Interestingly, we observed that L. donovani infection itself reduces the serum leptin levels in malnutrition. Exogenous leptin restored severe body weight loss and parasite load in the spleen and liver of malnourished infected mice compared to controls. Leptin increases functional CD8+ T-cell population, Granzyme-A expression down-regulates anergic T-cell markers such as PD-1 and CTLA-4. It was also noticed that, leptin suppresses GM-CSF mRNA expression in parasite favored monocytes and reduced arginase activity in bone marrow derived macrophage indicate macrophages dependent T-cell activation and proliferation. Leptin-induced IFN-γ, IL-2, and TNF-α cytokines in the culture supernatant of splenocytes upon soluble leishmanial antigen (SLA) stimulation and significantly up-regulates serum IgG2a titers, which help to generate Th1 immune response in VL. Furthermore, leptin induced a granulomatous response and restored L. donovani induced tissue degeneration in the liver. Altogether, our findings suggest the exogenous leptin can restore T cell mediated immunity in malnourished VL mice.
Collapse
|
31
|
Cytokines Are Markers of the Clostridium difficile-Induced Inflammatory Response and Predict Disease Severity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00037-17. [PMID: 28592627 DOI: 10.1128/cvi.00037-17] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/24/2017] [Indexed: 12/25/2022]
Abstract
The host immune response affects pathogen virulence in Clostridium difficile infection (CDI). Thus, cytokine responses to CDI likely are associated with disease initiation and progression. Understanding the molecular drivers of inflammation and biochemical markers of disease severity is important for developing novel therapies and predicting disease prognosis. In this study, we investigated cytokine production in patients with CDI and evaluated the potential of cytokines to serve as biomarkers for CDI and predictors of disease severity. The systemic cytokine profiles of 36 CDI patients (20 with severe disease) and 8 healthy donors and the toxin-induced cytokine profiles of peripheral blood mononuclear cells (PBMC) were determined. Further, we evaluated glucosyltransferase (GT) activity in regulation of toxin-induced cytokine expression. We found upregulation of the majority of measured cytokines (11/20, 55%) in CDI patients. Interleukin-1β (IL-1β), IL-6, IL-8, IL-17A, and IL-16 were the most upregulated. High serum levels of IL-2 and IL-15 were associated with a poor prognosis in CDI patients, whereas high levels of IL-5 and gamma interferon (IFN-γ) were associated with less severe disease. Both TcdA and TcdB were potent inducers of cytokine responses, as demonstrated by stimulation of a greater number and amount of cytokines. In addition to confirming prior reports on the role of IL-8, IL-1β, and IL-6 in CDI, our data suggest that IL-16 and IL-17A, as well as the IL-1β/Th17 axis, play a key role in driving inflammatory responses in CDI. A functional GT domain of C. difficile toxins was required for the induction of a majority of cytokines investigated.
Collapse
|
32
|
Pérez-Cruz M, Bello-Gil D, Costa C, Mañez R. Cytokine Profile Associated with Selective Removal of Natural Anti-αGal Antibodies in a Sepsis Model in Gal-KO Mice. BIOCHEMISTRY (MOSCOW) 2017; 82:205-212. [PMID: 28320304 DOI: 10.1134/s0006297917020122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Selective depletion of natural anti-Galα1-3Galβ1-4GlcNAc (so-called anti-αGal) antibodies is achieved in α1,3-galactosyltransferase knockout (Gal-KO) mice by administration of the soluble glycoconjugate of αGal GAS914. This molecule removed up to 90% of natural circulating anti-αGal antibodies without causing unspecific production of cytokines in wild-type (CBA) and Gal-KO mice. However, the removal of anti-αGal antibodies in Gal-KO mice with GAS914 in the context of sepsis after cecal ligation and puncture (CLP) was associated with a significant increase in the production of leptin, CXLC1, CXLC13, and TIMP-1 cytokines compared to vehicle (PBS)-treated controls. Despite the current lack of understanding of the underlying mechanism, our data suggest a putative role of natural anti-αGal antibodies in the regulation of some cytokines during sepsis.
Collapse
Affiliation(s)
- Magdiel Pérez-Cruz
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, 08908, Spain
| | | | | | | |
Collapse
|
33
|
Role of Eosinophils and Tumor Necrosis Factor Alpha in Interleukin-25-Mediated Protection from Amebic Colitis. mBio 2017; 8:mBio.02329-16. [PMID: 28246365 PMCID: PMC5347349 DOI: 10.1128/mbio.02329-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The parasite Entamoeba histolytica is a cause of diarrhea in infants in low-income countries. Previously, it was shown that tumor necrosis factor alpha (TNF-α) production was associated with increased risk of E. histolytica diarrhea in children. Interleukin-25 (IL-25) is a cytokine that is produced by intestinal epithelial cells that has a role in maintenance of gut barrier function and inhibition of TNF-α production. IL-25 expression was decreased in humans and in the mouse model of amebic colitis. Repletion of IL-25 blocked E. histolytica infection and barrier disruption in mice, increased gut eosinophils, and suppressed colonic TNF-α. Depletion of eosinophils with anti-Siglec-F antibody prevented IL-25-mediated protection. In contrast, depletion of TNF-α resulted in resistance to amebic infection. We concluded that IL-25 provides protection from amebiasis, which is dependent upon intestinal eosinophils and suppression of TNF-α. The intestinal epithelial barrier is important for protection from intestinal amebiasis. We discovered that the intestinal epithelial cytokine IL-25 was suppressed during amebic colitis in humans and that protection could be restored in the mouse model by IL-25 administration. IL-25 acted via eosinophils and suppressed TNF-α. This work illustrates a previously unrecognized pathway of innate mucosal immune response.
Collapse
|
34
|
Buonomo EL, Petri WA. The microbiota and immune response during Clostridium difficile infection. Anaerobe 2016; 41:79-84. [PMID: 27212111 PMCID: PMC5050085 DOI: 10.1016/j.anaerobe.2016.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/09/2016] [Accepted: 05/16/2016] [Indexed: 12/27/2022]
Abstract
Clostridium difficile is a gram-positive, spore forming anaerobe that infects the gut when the normal microbiota has been disrupted. C. difficile infection (CDI) is the most common cause of hospital acquired infection in the United States, and the leading cause of death due to gastroenteritis. Patients suffering from CDI have varying symptoms which range from mild diarrhea to pseudomembranous colitis and death. The involvement of the immune response to influence disease severity is just beginning to be investigated. There is evidence that the immune response can facilitate either protective or pathogenic phenotypes, suggesting it plays a multifaceted role during CDI. In addition to the immune response, the microbiota is pivotal in dictating the pathogenesis to CDI. A healthy microbiota effectively inhibits infection by restricting the ability of C. difficile to expand in the colon. Thus, understanding which immune mediators and components of the microbiota play beneficial roles during CDI will be important to future therapeutic developments. This review outlines how the microbiota can modulate specific immune mediators, such as IL-23 and others, to influence disease outcome.
Collapse
Affiliation(s)
- Erica L Buonomo
- Department of Microbiology, Immunology and Cancer, University of Virginia Charlottesville, VA, 22908, USA.
| | - William A Petri
- Department of Microbiology, Immunology and Cancer, University of Virginia Charlottesville, VA, 22908, USA; Department of Medicine, University of Virginia, USA; Department of Pathology, University of Virginia, USA.
| |
Collapse
|
35
|
Karrasch T, Schaeffler A. Adipokines and the role of visceral adipose tissue in inflammatory bowel disease. Ann Gastroenterol 2016; 29:424-438. [PMID: 27708507 PMCID: PMC5049548 DOI: 10.20524/aog.2016.0077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/22/2016] [Indexed: 12/20/2022] Open
Abstract
Recently, adipocytes have been recognized as actively participating in local and systemic immune responses via the secretion of peptides detectable in relevant levels in the systemic circulation, the so-called "adipo(cyto)kines". Multiple studies appearing within the last 10-15 years have focused on the possible impact of adipose tissue depots on inflammatory bowel disease (IBD). Consequently, various hypotheses regarding the role of different adipokines in inflammatory diseases in general and in intestinal inflammatory processes in particular have been developed and have been further refined in recent years. After a focused summary of the data reported concerning the impact of visceral adipose tissue on IBD, such as Crohn's disease and ulcerative colitis, our review focuses on recent developments indicating that adipocytes as part of the innate immune system actively participate in antimicrobial host defenses in the context of intestinal bacterial translocation, which are of utmost importance for the homeostasis of the whole organism. Modulators of adipose tissue function and regulators of adipokine secretion, as well as modifiers of adipocytic pattern recognition molecules, might represent future potential drug targets in IBD.
Collapse
Affiliation(s)
- Thomas Karrasch
- Department of Internal Medicine III, Giessen University Hospital, Germany
| | - Andreas Schaeffler
- Department of Internal Medicine III, Giessen University Hospital, Germany
| |
Collapse
|
36
|
Buonomo EL, Cowardin CA, Wilson MG, Saleh MM, Pramoonjago P, Petri WA. Microbiota-Regulated IL-25 Increases Eosinophil Number to Provide Protection during Clostridium difficile Infection. Cell Rep 2016; 16:432-443. [PMID: 27346351 PMCID: PMC4945404 DOI: 10.1016/j.celrep.2016.06.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 12/17/2022] Open
Abstract
Clostridium difficile infection (CDI) is the most common cause of hospital-acquired infection in the United States. Host susceptibility and the severity of infection are influenced by disruption of the microbiota and the immune response. However, how the microbiota regulate immune responses to mediate CDI outcome remains unclear. Here, we have investigated the role of the microbiota-linked cytokine IL-25 during infection. Intestinal IL-25 was suppressed during CDI in humans and mice. Restoration of IL-25 reduced CDI-associated mortality and tissue pathology even though equivalent levels of C. difficile bacteria and toxin remained in the gut. IL-25 protection was mediated by gut eosinophils, as demonstrated by an increase in intestinal eosinophils and a loss of IL-25 protection upon eosinophil depletion. These findings support a mechanism whereby the induction of IL-25-mediated eosinophilia can reduce host mortality during active CDI. This work may provide targets for future development of microbial or immune-based therapies.
Collapse
Affiliation(s)
- Erica L Buonomo
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA
| | - Carrie A Cowardin
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA
| | - Madeline G Wilson
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA
| | - Mahmoud M Saleh
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA
| | - Patcharin Pramoonjago
- Department of Pathology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA
| | - William A Petri
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA; Department of Medicine, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA; Department of Pathology, University of Virginia (UVA), Health Sciences Center, Charlottesville, VA 22908, USA.
| |
Collapse
|
37
|
Jose S, Madan R. Neutrophil-mediated inflammation in the pathogenesis of Clostridium difficile infections. Anaerobe 2016; 41:85-90. [PMID: 27063896 DOI: 10.1016/j.anaerobe.2016.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/04/2016] [Indexed: 12/19/2022]
Abstract
Clostridium difficile is the most important cause of nosocomial infectious diarrhea in the western world. C. difficile infections are a major healthcare burden with approximately 500,000 new cases every year and an estimated annual cost of nearly $1 billion in the U.S. Furthermore, the infections are no longer restricted to health care facilities, and recent studies indicate spread of C. difficile infection to the community as well. The clinical spectrum of C. difficile infection ranges from asymptomatic colonization to severe diarrhea, fulminant colitis and death. This spectrum results from a complex interplay between bacterial virulence factors, the colonic microbiome and the host inflammatory response. The overall vigor of host inflammatory response is believed to be an important determinant of C. difficile disease severity, and a more robust immune response is associated with worse outcomes. Neutrophils are the primary cells that respond to C. difficile invasion and neutrophilic inflammation is the hallmark of C. difficile-associated disease. In this review, we will focus on the role of neutrophils (infiltration to infected tissue, pathogen clearance and resolution of inflammation) in the immuno-pathogenesis of C. difficile-associated disease (CDAD).
Collapse
Affiliation(s)
- Shinsmon Jose
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, OH 45267, USA
| | - Rajat Madan
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati, OH 45267, USA.
| |
Collapse
|
38
|
Naylor C, Petri WA. Leptin Regulation of Immune Responses. Trends Mol Med 2016; 22:88-98. [PMID: 26776093 DOI: 10.1016/j.molmed.2015.12.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/06/2015] [Accepted: 12/10/2015] [Indexed: 12/19/2022]
Abstract
Leptin is a regulatory hormone with multiple roles in the immune system. We favor the concept that leptin signaling 'licenses' various immune cells to engage in immune responses and/or to differentiate. Leptin is an inflammatory molecule that is capable of activating both adaptive and innate immunity. It can also 'enhance' immune functions, including inflammatory cytokine production in macrophages, granulocyte chemotaxis, and increased Th17 proliferation. Leptin can also 'inhibit' cells; CD4(+) T cells are inhibited from differentiating into regulatory T cells in the presence of elevated leptin, while NK cells can exhibit impaired cytotoxicity under the same circumstances. Consequently, understanding the effect of leptin signaling is important to appreciate various aspects of immune dysregulation observed in malnutrition, obesity, and autoimmunity.
Collapse
Affiliation(s)
- Caitlin Naylor
- Medical Research Council Unit, Atlantic Blvd, Serrekunda, Gambia.
| | | |
Collapse
|
39
|
Madan R, Petri WA. Role of obesity and adipose tissue-derived cytokine leptin during Clostridium difficile infection. Anaerobe 2015; 34:182-6. [PMID: 25638400 DOI: 10.1016/j.anaerobe.2014.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 10/30/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023]
Abstract
Obesity is among the most pressing health concerns in the world since it is increasingly common even in the developing world, and is clearly associated with increased risk for chronic debilitating diseases and death. Furthermore, obesity can influence the pathogenesis of infectious diseases by affecting the balance of pathogen clearance and pathological inflammation. The mechanisms that result in enhanced inflammation in obese individuals are poorly understood. Clostridium difficile is a major cause of nosocomial infections worldwide. Recent studies have shown that obesity is associated with increased risk of C. difficile infections. In this review, we will discuss our current knowledge of the role of obesity in determining risk of C. difficile infections, and focus on the role of the adipose tissue-derived cytokine leptin in C. difficile infections.
Collapse
Affiliation(s)
- Rajat Madan
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Charlottesville, VA 22908, USA.
| | - William A Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, USA; Department of Pathology, University of Virginia, USA.
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW This review summarizes current knowledge on the contribution of mesenteric adipose tissue in intestinal inflammation. We will describe the cellular and humoral characteristics of creeping fat, their potential impact for Crohn's disease and propose a working model for the critical interplay between the creeping fat and the inflamed intestine. RECENT FINDINGS Creeping fat can be distinguished from healthy adipose tissue by its distinctively small adipocytes, by a specific microenvironment defined by high levels of adipokines and by a dominant immune cell infiltration. In Crohn's disease transmural inflammation facilitates increased bacterial translocation into the creeping fat. Translocalizing antigens can directly activate (pre)adipocytes via innate receptors. Adipocyte-derived mediators modulate phenotype and function of innate and adaptive immune cells. Activated (pre)adipocytes and adipokine-modulated immune cells might support a degree of inflammatory activation within the creeping fat that allows competent immune defense against exogenous factors while preventing systemic inflammation. SUMMARY Fat tissue as an active organ in health and disease has been ignored for too long. The last few years of research provided evidence for the complex metabolic and immunological functions of adipose tissue. On the basis of the available data, creeping fat in Crohn's disease exerts a protective function by a localized anti-inflammatory effect, thus preventing a systemic inflammatory response.
Collapse
|
41
|
Persistence and toxin production by Clostridium difficile within human intestinal organoids result in disruption of epithelial paracellular barrier function. Infect Immun 2014; 83:138-45. [PMID: 25312952 DOI: 10.1128/iai.02561-14] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile is the leading cause of infectious nosocomial diarrhea. The pathogenesis of C. difficile infection (CDI) results from the interactions between the pathogen, intestinal epithelium, host immune system, and gastrointestinal microbiota. Previous studies of the host-pathogen interaction in CDI have utilized either simple cell monolayers or in vivo models. While much has been learned by utilizing these approaches, little is known about the direct interaction of the bacterium with a complex host epithelium. Here, we asked if human intestinal organoids (HIOs), which are derived from pluripotent stem cells and demonstrate small intestinal morphology and physiology, could be used to study the pathogenesis of the obligate anaerobe C. difficile. Vegetative C. difficile, microinjected into the lumen of HIOs, persisted in a viable state for up to 12 h. Upon colonization with C. difficile VPI 10463, the HIO epithelium is markedly disrupted, resulting in the loss of paracellular barrier function. Since similar effects were not observed when HIOs were colonized with the nontoxigenic C. difficile strain F200, we directly tested the role of toxin using TcdA and TcdB purified from VPI 10463. We show that the injection of TcdA replicates the disruption of the epithelial barrier function and structure observed in HIOs colonized with viable C. difficile.
Collapse
|
42
|
Host recognition of Clostridium difficile and the innate immune response. Anaerobe 2014; 30:205-9. [PMID: 25223264 DOI: 10.1016/j.anaerobe.2014.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 08/12/2014] [Accepted: 08/28/2014] [Indexed: 02/08/2023]
Abstract
Clostridium difficile is a Gram-positive, spore forming bacillus and the most common cause of antibiotic-associated diarrhea in the United States. Clinical outcomes of C. difficile infection (CDI) range from asymptomatic colonization to pseudomembranous colitis, sepsis and death. Disease is primarily mediated by the action of the Rho-glucosylating toxins A and B, which induce potent pro-inflammatory signaling within the host. The role of this inflammatory response during infection is just beginning to be appreciated, with recent data suggesting inflammatory markers correlate closely with disease severity. In addition to the toxins, multiple innate immune signaling pathways have been implicated in establishing an inflammatory response during infection. In intoxication-based models of disease, inflammation typically enhances pathogenesis, while protection from infection seems to require some level of inflammatory response. Thus, the host immune response plays a key role in shaping the course of infection and a balanced inflammatory response which eradicates infection without damaging host tissues is likely required for successful resolution of disease.
Collapse
|
43
|
Mancuso P, O′Brien E, Prano J, Goel D, Aronoff DM. No Impairment in host defense against Streptococcus pneumoniae in obese CPEfat/fat mice. PLoS One 2014; 9:e106420. [PMID: 25203099 PMCID: PMC4159279 DOI: 10.1371/journal.pone.0106420] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/06/2014] [Indexed: 12/16/2022] Open
Abstract
In the US and globally, dramatic increases in the prevalence of adult and childhood obesity have been reported during the last 30 years. In addition to cardiovascular disease, type II diabetes, and liver disease, obesity has recently been recognized as an important risk factor for influenza pneumonia. During the influenza pandemic of 2009, obese individuals experienced a greater severity of illness from the H1N1 virus. In addition, obese mice have also been shown to exhibit increased lethality and aberrant pulmonary inflammatory responses following influenza infection. In contrast to influenza, the impact of obesity on bacterial pneumonia in human patients is controversial. In this report, we compared the responses of lean WT and obese CPEfat/fat mice following an intratracheal infection with Streptococcus pneumoniae, the leading cause of community-acquired pneumonia. At 16 weeks of age, CPEfat/fat mice develop severe obesity, hyperglycemia, elevated serum triglycerides and leptin, and increased blood neutrophil counts. There were no differences between lean WT and obese CPEfat/fat mice in survival or lung and spleen bacterial burdens following intratracheal infection with S. pneumoniae. Besides a modest increase in TNF-α levels and increased peripheral blood neutrophil counts in CPEfat/fat mice, there were not differences in lung or serum cytokines after infection. These results suggest that obesity, accompanied by hyperglycemia and modestly elevated triglycerides, at least in the case of CPEfat/fat mice, does not impair innate immunity against pneumococcal pneumonia.
Collapse
Affiliation(s)
- Peter Mancuso
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Edmund O′Brien
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Joseph Prano
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Deepti Goel
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - David M. Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
44
|
Abstract
Studies in human populations and mouse models of disease have linked the common leptin receptor Q223R mutation to obesity, multiple forms of cancer, adverse drug reactions, and susceptibility to enteric and respiratory infections. Contradictory results cast doubt on the phenotypic consequences of this variant. We set out to determine whether the Q223R substitution affects leptin binding kinetics using surface plasmon resonance (SPR), a technique that allows sensitive real-time monitoring of protein-protein interactions. We measured the binding and dissociation rate constants for leptin to the extracellular domain of WT and Q223R murine leptin receptors expressed as Fc-fusion proteins and found that the mutant receptor does not significantly differ in kinetics of leptin binding from the WT leptin receptor. (WT: ka 1.76×106±0.193×106 M−1 s−1, kd 1.21×10−4±0.707×10−4 s−1, KD 6.47×10−11±3.30×10−11 M; Q223R: ka 1.75×106±0.0245×106 M−1 s−1, kd 1.47×10−4±0.0505×10−4 s−1, KD 8.43×10−11±0.407×10−11 M). Our results support earlier findings that differences in affinity and kinetics of leptin binding are unlikely to explain mechanistically the phenotypes that have been linked to this common genetic variant. Future studies will seek to elucidate the mechanism by which this mutation influences susceptibility to metabolic, infectious, and malignant pathologies.
Collapse
|