1
|
Burge KY, Georgescu C, Zhong H, Wilson AP, Gunasekaran A, Yu Z, Franca A, Eckert JV, Wren JD, Chaaban H. Spatial transcriptomics delineates potential differences in intestinal phenotypes of cardiac and classical necrotizing enterocolitis. iScience 2025; 28:112166. [PMID: 40201118 PMCID: PMC11978348 DOI: 10.1016/j.isci.2025.112166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/20/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal gastrointestinal disease, often resulting in multi-organ failure and death. While classical NEC is strictly associated with prematurity, cardiac NEC is a subset of the disease occurring in infants with comorbid congenital heart disease. Despite similar symptomatology, the NEC subtypes vary slightly in presentation and may represent etiologically distinct diseases. We compared ileal spatial transcriptomes of patients with cardiac and classical NEC. Epithelial and immune cells cluster well by cell-type segment and NEC subtype. Differences in metabolism and immune cell activation functionally differentiate the cell-type makeup of the NEC subtypes. The classical NEC phenotype is defined by dysbiosis-induced inflammatory signaling and metabolic acidosis, while that of cardiac NEC involves reduced angiogenesis and endoplasmic reticulum stress-induced apoptosis. Despite subtype-associated clinical and demographic variability, spatial transcriptomics has substantiated pathway and network differences within immune and epithelial segments between cardiac and classical NEC.
Collapse
Affiliation(s)
- Kathryn Y. Burge
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Constantin Georgescu
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hua Zhong
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Adam P. Wilson
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Aarthi Gunasekaran
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zhongxin Yu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Addison Franca
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jeffrey V. Eckert
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jonathan D. Wren
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Hala Chaaban
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
2
|
Pasetti MF, Milletich PL, White JA, Butts J, Brady RC, Dickey MD, Ballou C, Maier N, Sztein MB, Baqar S, Louis Bourgeois A, Bernstein DI. Safety and immunogenicity in humans of enterotoxigenic Escherichia coli double mutant heat-labile toxin administered intradermally. NPJ Vaccines 2025; 10:23. [PMID: 39893179 PMCID: PMC11787345 DOI: 10.1038/s41541-025-01071-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) diarrhea is associated with a high burden of disease globally, for which no licensed vaccine is available. A Phase 1, double-blind, dose-escalation (0.1-2.0 µg) study was conducted to evaluate the safety and immunogenicity of double mutant heat-labile toxin LTR192G/L211A (dmLT) delivered intradermally (ID) to healthy adults. Subjects received up to three immunizations at three-week intervals. The vaccine was safe, although it induced mild local and some gastrointestinal adverse events, as well as frequent hyperpigmentation at the injection site. High levels of serum IgG and IgA, LT neutralizing antibodies, and IgG and IgA antibodies in lymphocyte supernatant were elicited post-vaccination, most prominently at the largest dose (2.0 μg). Rates of responses were the highest in subjects who received the largest dose (2.0 μg) and multiple immunizations. The ETEC dmLT vaccine was safe and highly immunogenic, inducing long-lasting systemic and mucosal responses when administered by the ID route. Trial registration Clinical Trials NCT02531685.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Patricia L Milletich
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Rebecca C Brady
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michelle D Dickey
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Nicole Maier
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - A Louis Bourgeois
- PATH, Washington, DC, USA
- John Hopkins University School of Public Health, Baltimore, MD, USA
| | - David I Bernstein
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Zimmermann W, Kammerer R. Evolution of CEACAM pathogen decoy receptors in primates. Eur J Clin Invest 2024; 54 Suppl 2:e14356. [PMID: 39674876 DOI: 10.1111/eci.14356] [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: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND CEACAM1 in leukocytes controls cell activation during inflammation. This and its expression in epithelial cells led to frequent independent appropriation of CEACAM1 as receptor by pathogens in humans and other species to gain host access and to downregulate its immune response. As a countermeasure, decoy receptors with CEACAM1-like pathogen-binding domains evolved. The granulocyte-specific human CEACAM3 endocytic receptor diverts CEACAM1-binding pathogens to neutrophils for internalization and destruction. The role of the glycosylphosphatidylinositol-anchored CEACAM5 and CEACAM6 which can also bind CEACAM1-targeting pathogens in humans is less clear. METHODS We analyzed the selection of CEACAMs to avoid pathogen binding and to maintain similarity between pathogen receptors and decoy receptors in 148 primate species. RESULTS Notably, functional CEACAM3 genes were not found in gibbons and New World monkeys. Interestingly, CEACAM6 in these primates exhibits similar high ratios of rates of nonsynonymous and synonymous substitution (dN/dS) in their pathogen-binding N domain exons as found for CEACAM1. High dN/dS ratios are indicative of selection for diversification typically seen in pathogen receptors. Human CEACAM6 is expressed on granulocytes and epithelial cells. Therefore, CEACAM6 could substitute for the missing endocytic receptor CEACAM3. In nearly all investigated primate groups also N exons of the epithelially expressed CEACAM5 exhibit selection for diversification. In African populations, five high-frequency polymorphisms are observed in the pathogen-binding region of CEACAM5 (I80V, V83A, I100T, I112V, I113T) with 3-4 polymorphisms combined in the same individual. These polymorphisms correspond to CEACAM1 pathogen-binding domain sequences. CONCLUSION The glycosylphosphatidylinositol-anchored CEACAM5 and CEACAM6 are under selection to maintain similarity to the pathogen receptor CEACAM1 in most primate species, indicating a function as decoy receptors.
Collapse
Affiliation(s)
- Wolfgang Zimmermann
- Tumor Immunology Laboratory, LIFE Center, Department of Urology, University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Robert Kammerer
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| |
Collapse
|
4
|
Fleckenstein JM, Najjar SM, Zimmermann W, Hauck CR, Nguyen Q, Mejias-Luque R, Bhattacharyya A, McCarthy AJ, Sarkar A, Kujawski M, Konieva A, Elyateem F, Kube-Golovin I, Wennemuth G, Kammerer R, Skubitz KM, Shively JE, Dery KJ, Dveksler G, Götz L, Kleefeldt F, Ergün S. Current investigation of carcinoembryonic antigen cell adhesion molecule (CEACAM) biology summary of the 32nd CEA symposium: 20-23 September 2024. Würzburg, Germany. Eur J Clin Invest 2024; 54 Suppl 2:e14355. [PMID: 39674873 PMCID: PMC11880994 DOI: 10.1111/eci.14355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 12/17/2024]
Affiliation(s)
| | - Sonia M. Najjar
- Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | | | | | | | | | - Asima Bhattacharyya
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, Bhubaneswar, India
| | | | - Arup Sarkar
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, Bhubaneswar, India
| | | | | | | | | | | | - Robert Kammerer
- Friedrich-Loeffler Institut, Greifswald – Insel Riems, Germany
| | | | | | - Kenneth J. Dery
- University of California, Los Angeles, Los Angeles, California, USA
| | - Gabriela Dveksler
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Lisa Götz
- Julius Maximilians Universität, Würzburg, Germany
| | | | | |
Collapse
|
5
|
Sheikh A, Ganguli D, Vickers TJ, Singer BB, Foulke-Abel J, Akhtar M, Khatoon N, Setu B, Basu S, Harro C, Maier N, Beatty WL, Chakraborty S, Bhuiyan TR, Qadri F, Donowitz M, Fleckenstein JM. Host-derived CEACAM-laden vesicles engage enterotoxigenic Escherichia coli for elimination and toxin neutralization. Proc Natl Acad Sci U S A 2024; 121:e2410679121. [PMID: 39264739 PMCID: PMC11420188 DOI: 10.1073/pnas.2410679121] [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: 05/28/2024] [Accepted: 08/10/2024] [Indexed: 09/14/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of diarrheal illnesses annually ranging from mildly symptomatic cases to severe, life-threatening cholera-like diarrhea. Although ETEC are associated with long-term sequelae including malnutrition, the acute diarrheal illness is largely self-limited. Recent studies indicate that in addition to causing diarrhea, the ETEC heat-labile toxin (LT) modulates the expression of many genes in intestinal epithelia, including carcinoembryonic cell adhesion molecules (CEACAMs) which ETEC exploit as receptors, enabling toxin delivery. Here, however, we demonstrate that LT also enhances the expression of CEACAMs on extracellular vesicles (EV) shed by intestinal epithelia and that CEACAM-laden EV increase in abundance during human infections, mitigate pathogen-host interactions, scavenge free ETEC toxins, and accelerate ETEC clearance from the gastrointestinal tract. Collectively, these findings indicate that CEACAMs play a multifaceted role in ETEC pathogen-host interactions, transiently favoring the pathogen, but ultimately contributing to innate responses that extinguish these common infections.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Debayan Ganguli
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Tim J. Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Bernhard B. Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, 45147Essen, Germany
| | - Jennifer Foulke-Abel
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Marjahan Akhtar
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
- Enteric and Respiratory. Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka1212, Bangladesh
| | - Nazia Khatoon
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Bipul Setu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
| | - Clayton Harro
- Division of Global Disease Epidemiology and Control with the Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD21205
| | - Nicole Maier
- Center for Vaccine Innovation and Access, PATH, Seattle, WA98121
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Subhra Chakraborty
- Division of Global Disease Epidemiology and Control with the Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD21205
| | - Taufiqur R. Bhuiyan
- Enteric and Respiratory. Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka1212, Bangladesh
| | - Firdausi Qadri
- Enteric and Respiratory. Infections, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka1212, Bangladesh
| | - Mark Donowitz
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - James M. Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, MO63110
- Medicine Service, Infectious Disease Section, Veterans Affairs Health Care System, Saint Louis, MO63106
| |
Collapse
|
6
|
Sheikh A, Ganguli D, Vickers TJ, Singer B, Foulke-Abel J, Akhtar M, Khatoon N, Setu B, Basu S, Harro C, Maier N, Beatty WL, Chakraborty S, Bhuiyan TR, Qadri F, Donowitz M, Fleckenstein JM. Host-derived CEACAM-laden vesicles engage enterotoxigenic E. coli for elimination and toxin neutralization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604983. [PMID: 39091797 PMCID: PMC11291149 DOI: 10.1101/2024.07.24.604983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of diarrheal illnesses annually ranging from mildly symptomatic cases to severe, life-threatening cholera-like diarrhea. Although ETEC are associated with long-term sequelae including malnutrition, the acute diarrheal illness is largely self-limited. Recent studies indicate that in addition to causing diarrhea, the ETEC heat-labile toxin (LT) modulates the expression of many genes in intestinal epithelia, including carcinoembryonic cell adhesion molecules (CEACAMs) which ETEC exploit as receptors, enabling toxin delivery. Here however, we demonstrate that LT also enhances the expression of CEACAMs on extracellular vesicles (EV) shed by intestinal epithelia and that CEACAM-laden EV increase in abundance during human infections, mitigate pathogen-host interactions, scavenge free ETEC toxins, and accelerate ETEC clearance from the gastrointestinal tract. Collectively, these findings indicate that CEACAMs play a multifaceted role in ETEC pathogen-host interactions, transiently favoring the pathogen, but ultimately contributing to innate responses that extinguish these common infections.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Debayan Ganguli
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Tim J. Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Bernhard Singer
- Institute of Anatomy, Medical Faculty, University of Suisberg-Essen, 45147 Essen, Germany
| | - Jennifer Foulke-Abel
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marjahan Akhtar
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
- International Centre for Diarrhoeal Disease Research, Bangladesh; Dhaka, Bangladesh
| | - Nazia Khatoon
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Bipul Setu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
| | - Clayton Harro
- Department of International Health, Division of Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health
| | | | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Subhra Chakraborty
- Department of International Health, Division of Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health
| | - Tafiqur R. Bhuiyan
- International Centre for Diarrhoeal Disease Research, Bangladesh; Dhaka, Bangladesh
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh; Dhaka, Bangladesh
| | - Mark Donowitz
- Division of Gastroenterology & Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James M. Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, USA
- Medicine Service, Infectious Disease Section, Veterans Affairs Health Care System, Saint Louis, Missouri, USA
| |
Collapse
|
7
|
Sabui S, Anthonymuthu S, Ramamoorthy K, Skupsky J, Jennings TSK, Rahmatpanah F, Fleckenstein JM, Said HM. Effect of knocking out mouse Slc44a4 on colonic uptake of the microbiota-generated thiamine pyrophosphate and colon physiology. Am J Physiol Gastrointest Liver Physiol 2024; 327:G36-G46. [PMID: 38713615 PMCID: PMC11376973 DOI: 10.1152/ajpgi.00065.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/09/2024]
Abstract
Humans and mammals obtain vitamin B1 from dietary and gut microbiota sources. A considerable amount of the microbiota-generated vitamin exists in the form of thiamine pyrophosphate (TPP), and colonocytes are capable of absorbing TPP via a specific carrier-mediated process that involves the colonic TPP transporter (cTPPT encoded by SLC44A4). Little is known about the relative contribution of the SLC44A4 transporter toward total colonic carrier-mediated TPP uptake and its role in colon physiology. To address these issues, we generated an Slc44a4 knockout (KO) mouse model (by Cre-Lox recombination) and found a near-complete inhibition in colonic carrier-mediated [3H]TPP uptake in the Slc44a4 KO compared with wild-type (WT) littermates. We also observed a significant reduction in KO mice's body weight and a shortening of their colon compared with WT. Using RNAseq and Ingenuity pathway analysis (IPA) approaches, we found that knocking out the colonic Slc44a4 led to changes in the level of expression of many genes, including upregulation in those associated with intestinal inflammation and colitis. Finally, we found that the Slc44a4 KO mice were more susceptible to the effect of the colitogenic dextran sodium sulfate (DSS) compared with WT animals, a finding that lends support to the recent prediction by multiple genome-wide association studies (GWAS) that SLC44A4 is a possible colitis susceptibility gene. In summary, the results of these investigations show that Slc44a4 is the predominant or only transporter involved in the colonic uptake of TPP, that the transporter is important for colon physiology, and that its deletion increases susceptibility to inflammation.NEW & NOTEWORTHY This study shows that Slc44a4 is the predominant or only transport system involved in the uptake of the gut microbiota-generated thiamine pyrophosphate (TPP) in the colon and that its deletion affects colon physiology and increases its susceptibility to inflammation.
Collapse
Affiliation(s)
- Subrata Sabui
- Department of Physiology and Biophysics, University of California, Irvine, California, United States
- Veterans Affairs Medical Center, Long Beach, California, United States
| | - Selvaraj Anthonymuthu
- Department of Physiology and Biophysics, University of California, Irvine, California, United States
| | - Kalidas Ramamoorthy
- Department of Physiology and Biophysics, University of California, Irvine, California, United States
| | - Jonathan Skupsky
- Veterans Affairs Medical Center, Long Beach, California, United States
- Department of Medicine, University of California, Irvine, California, United States
| | - Tara Sinta Kartika Jennings
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, United States
| | - Farah Rahmatpanah
- Department of Pathology and Laboratory Medicine, University of California, Irvine, California, United States
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Veterans Affairs Medical Center, St. Louis, Missouri, United States
| | - Hamid M Said
- Department of Physiology and Biophysics, University of California, Irvine, California, United States
- Veterans Affairs Medical Center, Long Beach, California, United States
- Department of Medicine, University of California, Irvine, California, United States
| |
Collapse
|
8
|
Ayibieke A, Wajima T, Kano S, Chatterjee NS, Hamabata T. The colonization factor CS6 of enterotoxigenic Escherichia coli contributes to host cell invasion. Microb Pathog 2024; 190:106636. [PMID: 38556103 DOI: 10.1016/j.micpath.2024.106636] [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: 12/27/2023] [Revised: 03/11/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the main causes of diarrhea in children and travelers in low-income regions. The virulence of ETEC is attributed to its heat-labile and heat-stable enterotoxins, as well as its colonization factors (CFs). CFs are essential for ETEC adherence to the intestinal epithelium. However, its invasive capability remains unelucidated. In this study, we demonstrated that the CS6-positive ETEC strain 4266 can invade mammalian epithelial cells. The invasive capability was reduced in the 4266 ΔCS6 mutant but reintroduction of CS6 into this mutant restored the invasiveness. Additionally, the laboratory E. coli strain Top 10, which lacks the invasive capability, was able to invade Caco-2 cells after gaining the CS6-expressing plasmid pCS6. Cytochalasin D inhibited cell invasion in both 4266 and Top10 pCS6 cells, and F-actin accumulation was observed near the bacteria on the cell membrane, indicating that CS6-positive bacteria were internalized via actin polymerization. Other cell signal transduction inhibitors, such as genistein, wortmannin, LY294002, PP1, and Ro 32-0432, inhibited the CS6-mediated invasion of Caco-2 cells. The internalized bacteria of both 4266 and Top10 pCS6 strains were able to survive for up to 48 h, and 4266 cells were able to replicate within Caco-2 cells. Immunofluorescence microscopy revealed that the internalized 4266 cells were present in bacteria-containing vacuoles, which underwent a maturation process indicated by the recruitment of the early endosomal marker EEA-1 and late endosomal marker LAMP-1 throughout the infection process. The autophagy marker LC3 was also observed near these vacuoles, indicating the initiation of LC-3-associated phagocytosis (LAP). However, intracellular bacteria continued to replicate, even after the initiation of LAP. Moreover, intracellular filamentation was observed in 4266 cells at 24 h after infection. Overall, this study shows that CS6, in addition to being a major CF, mediates cell invasion. This demonstrates that once internalized, CS6-positive ETEC is capable of surviving and replicating within host cells. This capability may be a key factor in the extended and recurrent nature of ETEC infections in humans, thus highlighting the critical role of CS6.
Collapse
Affiliation(s)
- Alafate Ayibieke
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takeaki Wajima
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shigeyuki Kano
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | - Takashi Hamabata
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
9
|
Kovanda L, Rengman S, Tawde S, Pos J, Park S, Sun S, Park J, Kim K, Li X, Liu Y. Dietary glycerides of valerate ameliorate diarrhea and impact intestinal physiology and serum biomarkers in weaned piglets infected with enterotoxigenic Escherichia coli F18. J Anim Sci 2024; 102:skae322. [PMID: 39432563 PMCID: PMC11537800 DOI: 10.1093/jas/skae322] [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: 05/31/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
In the commercial swine farm setting, the postweaning period is a critical window during which piglets are highly susceptible to infection and enterotoxigenic E. coli (ETEC)-associated diarrhea. Short-chain fatty acids and their glycerides are compounds that may influence intestinal health; however, valerate is one that has not been well-characterized for its role as a dietary supplement. Therefore, the major objective of this experiment was to investigate two forms of valerate glycerides on diarrhea, intestinal physiology, and systemic immunity of weaned pigs experimentally infected with ETEC F18. Dietary treatments included a control diet and three additional diets supplemented with 0.075% monovalerin, 0.1% monovalerin, or 0.1% trivalerin, respectively. Piglets were weaned (21 d to 24 d of age), individually housed, and experimental diets were fed through the 28-d trial period. After a 7-d period, all piglets were inoculated on three consecutive days with 1010 CFU ETEC F18/3 mL. Growth performance was monitored throughout the trial, and daily diarrhea scores were recorded. Rectal swabs were collected for bacterial culture to confirm the presence or absence of β-hemolytic coliforms throughout the trial. Serum samples were collected and analyzed for inflammatory biomarkers on days 0, 3, 6, and 21 postinoculation (PI) and untargeted metabolomics on day 6 PI. Intestinal mucosa and tissue sections were harvested from pigs sacrificed on day 7 PI for gene expression and histology analysis. All data, except for frequency of diarrhea and metabolomics, were analyzed by ANOVA using the PROC MIXED of SAS. Dietary trivalerin reduced (P < 0.05) the frequency of severe diarrhea over the entire trial period and the frequency of β-hemolytic coliforms on day 7 PI compared with the control. The intestinal villus height on day 7 PI in jejunum tissue was increased (P < 0.05) in pigs fed trivalerin. The mRNA expression of TNF-α was decreased (P < 0.05) in the trivalerin group, while that of ZO1 was increased (P < 0.05) compared with control. Throughout the trial, serum TNF-α was reduced in pigs fed trivalerin compared with control. Serum metabolites, adenosine, inosine, and shikimic acid were reduced (P < 0.05) on day 6 PI in all treatment groups compared with control. In conclusion, the present results indicate supplementing dietary valerate glycerides exhibited beneficial impacts on diarrhea, inflammation, and intestinal gene expression of piglets during the postweaning period.
Collapse
Affiliation(s)
- Lauren Kovanda
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | - Sofia Rengman
- Department of Population Health and Reproduction, Perstorp Animal Nutrition, Waspik, The Netherlands
| | - Snehal Tawde
- Department of Population Health and Reproduction, Perstorp Animal Nutrition, Waspik, The Netherlands
| | - Jeroen Pos
- Department of Population Health and Reproduction, Perstorp Animal Nutrition, Waspik, The Netherlands
| | - Sangwoo Park
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | - Shuhan Sun
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | - Jungjae Park
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Kwangwook Kim
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Xunde Li
- School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA 95616, USA
| |
Collapse
|
10
|
Porter CK, Talaat KR, Isidean SD, Kardinaal A, Chakraborty S, Gutiérrez RL, Sack DA, Bourgeois AL. The Controlled Human Infection Model for Enterotoxigenic Escherichia coli. Curr Top Microbiol Immunol 2024; 445:189-228. [PMID: 34669040 DOI: 10.1007/82_2021_242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The controlled human infection model (CHIM) for enterotoxigenic Escherichia coli (ETEC) has been instrumental in defining ETEC as a causative agent of acute watery diarrhea, providing insights into disease pathogenesis and resistance to illness, and enabling preliminary efficacy evaluations for numerous products including vaccines, immunoprophylactics, and drugs. Over a dozen strains have been evaluated to date, with a spectrum of clinical signs and symptoms that appear to replicate the clinical illness seen with naturally occurring ETEC. Recent advancements in the ETEC CHIM have enhanced the characterization of clinical, immunological, and microbiological outcomes. It is anticipated that omics-based technologies applied to ETEC CHIMs will continue to broaden our understanding of host-pathogen interactions and facilitate the development of primary and secondary prevention strategies.
Collapse
Affiliation(s)
- Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA.
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Sandra D Isidean
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation, Bethesda, MD, 20817, USA
| | - Alwine Kardinaal
- NIZO Food Research, Ede, P.O. Box 20, 6710 BA EDE, Kernhemseweg 2, 6718 ZB EDE, The Netherlands
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Ramiro L Gutiérrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - David A Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - A Louis Bourgeois
- PATH|Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Suite 1000, Washington, DC, 20001, USA
| |
Collapse
|
11
|
Anthonymuthu S, Sabui S, Lee K, Sheikh A, Fleckenstein JM, Said HM. Bacterial lipopolysaccharide inhibits colonic carrier-mediated uptake of thiamin pyrophosphate: roles for TLR4 receptor and NF-κB/P38/JNK signaling pathway. Am J Physiol Cell Physiol 2023; 325:C758-C769. [PMID: 37519229 PMCID: PMC10635650 DOI: 10.1152/ajpcell.00272.2023] [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: 06/22/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023]
Abstract
This study investigated the effect of the bacterial endotoxin lipopolysaccharide (LPS) on colonic uptake of thiamin pyrophosphate (TPP), the biologically active form of vitamin B1 that is generated by gut microbiota. We used three complementary models in our study: in vitro (human-derived colonic epithelial NCM460), ex vivo (human differentiated colonoid monolayers), and in vivo (mouse colonic tissue). The results showed that exposure of NCM460 cells to LPS leads to a significant inhibition of carrier-mediated TPP uptake as well as in decreased expression of the colonic TPP transporter (cTPPT) protein, mRNA, and heterologous nuclear RNA (hnRNA) compared with untreated controls. Similarly, exposure of human differentiated colonoid monolayers and mice to LPS caused significant inhibition in colonic carrier-mediated TPP uptake and in cTPPT protein, mRNA, and hnRNA expression. The effect of LPS on colonic TPP uptake and cTTPT expression was also found to be associated with a significant reduction in activity of the SLC44A4 promoter as well as in decreased expression of the nuclear factor Elf-3 (E74-like ETS transcription factor 3), which is needed for promoter activity. Finally, we found that knocking down the Toll-like receptor 4 (TLR4) and blocking the nuclear factor kappa B (NF-κB), JNK, and p38 signaling pathways with the use of pharmacological inhibitors lead to significant abrogation in the degree of LPS-mediated inhibition in TPP uptake and cTPPT expression. These results demonstrated that exposure of colonic epithelia to LPS inhibits colonic TPP uptake via transcriptional mechanism(s) and that the effect is mediated via TLR4 receptor and NF-κB/p38/JNK signaling pathways.NEW & NOTEWORTHY This study examined the effect of the bacterial lipopolysaccharide (LPS) on the colonic uptake of thiamin pyrophosphate (TPP), the biologically active form of vitamin B1. Three complementary models were used: in vitro (human NCM460 cells), ex vivo (human colonoids), and in vivo (mice). The results showed LPS to significantly suppress TPP uptake and the expression of its transporter, and that these effects are mediated via the membrane TLR4 receptor, and involve the NF-κB/p38/JNK signaling pathways.
Collapse
Affiliation(s)
- Selvaraj Anthonymuthu
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Subrata Sabui
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| | - Katherine Lee
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Veterans Affairs Medical Center, St. Louis, Missouri, United States
| | - Hamid M Said
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medicine, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| |
Collapse
|
12
|
Sheikh A, Fleckenstein JM. Interactions of pathogenic Escherichia coli with CEACAMs. Front Immunol 2023; 14:1120331. [PMID: 36865539 PMCID: PMC9971599 DOI: 10.3389/fimmu.2023.1120331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
The pathogenic Escherichia coli can be parsed into specific variants (pathovars) depending on their phenotypic behavior and/or expression of specific virulence factors. These pathogens are built around chromosomally-encoded core attributes and through acquisition of specific virulence genes that direct their interaction with the host. Engagement of E. coli pathovars with CEACAMs is determined both by core elements common to all E. coli as well as extrachromosomally-encoded pathovar-specific virulence traits, which target amino terminal immunoglobulin variable-like (IgV) regions of CEACAMs. Emerging data suggests that engagement of CEACAMs does not unilaterally benefit the pathogen and that these interactions may also provide an avenue for pathogen elimination.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - James M. Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Infectious Diseases, Medicine Service, Veterans Affairs Saint Louis Health Care System, Saint Louis, MO, United States
| |
Collapse
|
13
|
Hollifield IE, Motyka NI, Stewart SR, Blyth MD, Fernando KA, Clement KL, Bitoun JP. Heat-Stable Enterotoxin Secretions Assessed via ICP-MS Reveal Iron-Mediated Regulation of Virulence in CFA/I- and CS6-Expressing ETEC Isolates. Cells 2023; 12:567. [PMID: 36831233 PMCID: PMC9954033 DOI: 10.3390/cells12040567] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/11/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are a significant cause of childhood diarrhea in low-resource settings. ETEC are defined by the production of heat-stable enterotoxin (ST) and/or heat-labile enterotoxin (LT), which alter intracellular cyclic nucleotide signaling and cause the secretion of water and electrolytes into the intestinal lumen. ETEC take cues from chemicals (e.g., glycans, bile salts, and solutes) that may be liberated following enterotoxin activity to recognize entrance into the host. ETEC then alter the expression of surface adhesins called colonization factors (CFs) to attach to the intestinal epithelium, proliferate, and cause disease. Here, we used an in vivo model of oral ST intoxication to determine its impact on luminal ion concentrations via ICP-MS. We also used functional assays, including Western blots, qPCR, and toxin activity assays, to assess the impact of luminal ion flux on CF and toxin expression. Finally, we assessed ETEC strains with CFs CFA/I or CS6 in a streptomycin mouse model of ETEC colonization. ST causes rapid and significant increases in luminal chloride but significant decreases in luminal magnesium and iron. We confirmed that increased sodium chloride suppresses CFA/I production in ETEC H10407 but does not affect CS6 production in ETEC 214-4. CFA/I production in ETEC H10407 is increased when magnesium becomes limiting, although it does not affect CS6 production in ETEC 214-4. Iron restriction via deferoxamine induces CFA/I expression in ETEC H10407 but not CS6 expression in ETEC 214-4. We demonstrate that ST production is suppressed via iron restriction in H10407, 214-4, and over 50 other ETEC clinical isolates. Lastly, we demonstrate that the iron restriction of mice using oral deferoxamine pre-treatment extends the duration of ETEC H10407 (CFA/I+) fecal shedding while accelerating ETEC 214-4 (CS6+) fecal shedding. Combined, these data suggest that enterotoxins modulate luminal ion flux to influence ETEC virulence including toxin and CF production.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jacob P. Bitoun
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
14
|
CEACAMS 1, 5, and 6 in disease and cancer: interactions with pathogens. Genes Cancer 2023; 14:12-29. [PMID: 36741860 PMCID: PMC9891707 DOI: 10.18632/genesandcancer.230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
The CEA family comprises 18 genes and 11 pseudogenes located at chromosome 19q13.2 and is divided into two main groups: cell surface anchored CEA-related cell adhesion molecules (CEACAMs) and the secreted pregnancy-specific glycoproteins (PSGs). CEACAMs are highly glycosylated cell surface anchored, intracellular, and intercellular signaling molecules with diverse functions, from cell differentiation and transformation to modulating immune responses associated with infection, inflammation, and cancer. In this review, we explore current knowledge surrounding CEACAM1, CEACAM5, and CEACAM6, highlight their pathological significance in the areas of cancer biology, immunology, and inflammatory disease, and describe the utility of murine models in exploring questions related to these proteins.
Collapse
|
15
|
Anthonymuthu S, Sabui S, Sheikh A, Fleckenstein JM, Said HM. Tumor necrosis factor α impedes colonic thiamin pyrophosphate and free thiamin uptake: involvement of JNK/ERK 1/2-mediated pathways. Am J Physiol Cell Physiol 2022; 323:C1664-C1680. [PMID: 36342158 PMCID: PMC9744649 DOI: 10.1152/ajpcell.00458.2022] [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: 10/12/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
The aim of this study was to examine the effect of TNFα (i.e., a predominant proinflammatory cytokine produced during chronic gut inflammation) on colonic uptake of thiamin pyrophosphate (TPP) and free thiamin, forms of vitamin B1 that are produced by the gut microbiota and are absorbed via distinct carrier-mediated systems. We utilized human-derived colonic epithelial CCD841 and NCM460 cells, human differentiated colonoid monolayers, and mouse intact colonic tissue preparations together with an array of cellular/molecular approaches in our investigation. The results showed that exposure of colonic epithelial cells to TNFα leads to a significant inhibition in TPP and free thiamin uptake. This inhibition was associated with: 1) a significant suppression in the level of expression of the colonic TPP transporter (cTPPT; encoded by SLC44A4), as well as thiamin transporters-1 & 2 (THTR-1 & -2; encoded by SLC19A2 & SLC19A3, respectively); 2) marked inhibition in activity of the SLC44A4, SLC19A2, and SLC19A3 promoters; and 3) significant suppression in level of expression of nuclear factors that are needed for activity of these promoters (i.e., CREB-1, Elf-3, NF-1A, SP-1). Furthermore, the inhibitory effects were found to be mediated via JNK and ERK1/2 signaling pathways. We also examined the level of expression of cTPPT and THTR-1 & -2 in colonic tissues of patients with active ulcerative colitis and found the levels to be significantly lower than in healthy controls. These findings demonstrate that exposure of colonocytes to TNFα suppresses TPP and free thiamin uptake at the transcriptional level via JNK- and Erk1/2-mediated pathways.
Collapse
Affiliation(s)
- Selvaraj Anthonymuthu
- Department of Physiology and Biophysics, University of California, Irvine, California
| | - Subrata Sabui
- Department of Physiology and Biophysics, University of California, Irvine, California
- Department of Medicine, University of California, Irvine, California
- Department of Medical Research, VA Medical Center, Long Beach, California
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Veterans Affairs Medical Center, St. Louis, Missouri
| | - Hamid M Said
- Department of Physiology and Biophysics, University of California, Irvine, California
- Department of Medicine, University of California, Irvine, California
- Department of Medical Research, VA Medical Center, Long Beach, California
| |
Collapse
|
16
|
Peng B, Cui Q, Ma C, Yi H, Gong P, Lin K, Liu T, Zhang L. Lactiplantibacillus plantarum YZX28 alleviated intestinal barrier dysfunction induced by enterotoxigenic Escherichia coli via inhibiting its virulence factor production. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Sheikh A, Tumala B, Vickers TJ, Martin JC, Rosa BA, Sabui S, Basu S, Simoes RD, Mitreva M, Storer C, Tyksen E, Head RD, Beatty W, Said HM, Fleckenstein JM. Enterotoxigenic Escherichia coli heat-labile toxin drives enteropathic changes in small intestinal epithelia. Nat Commun 2022; 13:6886. [PMID: 36371425 PMCID: PMC9653437 DOI: 10.1038/s41467-022-34687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/01/2022] [Indexed: 11/14/2022] Open
Abstract
Enterotoxigenic E. coli (ETEC) produce heat-labile (LT) and/or heat-stable (ST) enterotoxins, and commonly cause diarrhea in resource-poor regions. ETEC have been linked repeatedly to sequelae in children including enteropathy, malnutrition, and growth impairment. Although cellular actions of ETEC enterotoxins leading to diarrhea are well-established, their contributions to sequelae remain unclear. LT increases cellular cAMP to activate protein kinase A (PKA) that phosphorylates ion channels driving intestinal export of salt and water resulting in diarrhea. As PKA also modulates transcription of many genes, we interrogated transcriptional profiles of LT-treated intestinal epithelia. Here we show that LT significantly alters intestinal epithelial gene expression directing biogenesis of the brush border, the major site for nutrient absorption, suppresses transcription factors HNF4 and SMAD4 critical to enterocyte differentiation, and profoundly disrupts microvillus architecture and essential nutrient transport. In addition, ETEC-challenged neonatal mice exhibit substantial brush border derangement that is prevented by maternal vaccination with LT. Finally, mice repeatedly challenged with toxigenic ETEC exhibit impaired growth recapitulating the multiplicative impact of recurring ETEC infections in children. These findings highlight impacts of ETEC enterotoxins beyond acute diarrheal illness and may inform approaches to prevent major sequelae of these common infections including malnutrition that impact millions of children.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Brunda Tumala
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tim J Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John C Martin
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Subrata Sabui
- Departments of Medicine and Physiology/Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, 92697, USA
- Department of Research, VA Medical Center, Long Beach, CA, 90822, USA
| | - Supratim Basu
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rita D Simoes
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chad Storer
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Erik Tyksen
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Richard D Head
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Hamid M Said
- Departments of Medicine and Physiology/Biophysics, School of Medicine, University of California-Irvine, Irvine, CA, 92697, USA
- Department of Research, VA Medical Center, Long Beach, CA, 90822, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Infectious Diseases, Medicine Service, Veterans Affairs Saint Louis Health Care System, Saint Louis, MO, 63106, USA.
| |
Collapse
|
18
|
Activation of CEACAM1 with an agonistic monoclonal antibody results in inhibition of melanoma cells. Cancer Gene Ther 2022; 29:1676-1685. [PMID: 35681020 DOI: 10.1038/s41417-022-00486-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/08/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023]
Abstract
Inhibitory receptors (IRs), such as the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), are cell surface molecules expressed on both normal epithelial, endothelial, and hematopoietic cells and on neoplastic cells. IRs are usually used by cancer cells to inhibit immune cell functions. Thus, CEACAM1 positive tumor cells can interact homophilically with CEACAM1 expressed on T and NK cells to inhibit their antibody-dependent cell-mediated cytotoxicity (ADCC). In this study, we investigated the effect of agonistic/activating anti-CEACAM1 monoclonal antibody (mAb) on melanoma cell lines in vitro and in vivo, following our hypothesis that activation of CEACAM1 on melanoma cells by distinct mAbs may induce inhibition of cancer cell proliferation and/or their death. To address this, we established an activating anti-CEACAM1 mAb (CCM5.01) and characterized its binding to the CEACAM1 receptor. Using this mAb, we assessed the expression of CEACAM1 on four different human melanoma cell lines by western blot and flow cytometry and determined its effect on cell viability in vitro by MTT assay. Furthermore, we evaluated the mAb mechanism of action and found that binding of CEACAM1 with CCM5.01 induced SHP1 phosphorylation and p53 activation resulting in melanoma cell apoptosis. For in vivo studies, a xenograft model of melanoma was performed by injection of Mel-14 cells subcutaneously (s.c.) in SCID/Beige mice followed by intraperitoneal (i.p.) injection of CCM5.01 or of IgG1 isotype control every other day. CCM5.01 treated mice showed a slight but not significant decrease in tumor weight in comparison to the control group. Based on the obtained data, we suggest that activating CEACAM1 on melanoma cells might be a promising novel approach to fight cancers expressing this IR.
Collapse
|
19
|
Heat-labile enterotoxin enhances F4-producing enterotoxigenic E. coli adhesion to porcine intestinal epithelial cells by upregulating bacterial adhesins and STb enterotoxin. Vet Res 2022; 53:88. [PMID: 36303242 PMCID: PMC9615205 DOI: 10.1186/s13567-022-01110-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
As one of the crucial enterotoxins secreted by enterotoxigenic Escherichia coli (ETEC), heat-labile enterotoxin (LT) enhances bacterial adherence both in vivo and in vitro; however, the underlying mechanism remains unclear. To address this, we evaluated the adherence of LT-producing and LT-deficient ETEC strains using the IPEC-J2 cell model. The expression levels of inflammatory cytokines and chemokines, and tight-junction proteins were evaluated in IPEC-J2 cells after infection with various ETEC strains. Further, the levels of adhesins and enterotoxins were also evaluated in F4ac-producing ETEC (F4 + ETEC) strains after treatment with cyclic AMP (cAMP). The adherence of the ΔeltAB mutant was decreased compared with the wild-type strain, whereas adherence of the 1836-2/pBR322-eltAB strain was markedly increased compared with the 1836-2 parental strain. Production of LT up-regulated the expression of TNF-α, IL-6, CXCL-8, and IL-10 genes. However, it did not appear to affect tight junction protein expression. Importantly, we found that cAMP leads to the upregulation of adhesin production and STb enterotoxin. Moreover, the F4 + ETEC strains treated with cAMP also had greater adhesion to IPEC-J2 cells, and the adherence of ΔfaeG, ΔfliC, and ΔestB mutants was decreased. These results indicate that LT enhances the adherence of F4 + ETEC due primarily to the upregulation of F4 fimbriae, flagellin, and STb enterotoxin expression and provide insights into the pathogenic mechanism of LT and ETEC.
Collapse
|
20
|
Xiao K, Yang Y, Zhang Y, Lv Q, Huang F, Wang D, Zhao J, Liu Y. Long-chain PUFA ameliorate enterotoxigenic Escherichia coli-induced intestinal inflammation and cell injury by modulating pyroptosis and necroptosis signaling pathways in porcine intestinal epithelial cells. Br J Nutr 2022; 128:835-850. [PMID: 34915950 DOI: 10.1017/s0007114521005092] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study was aimed to investigate whether EPA and arachidonic acid (ARA), the representative n-3 or n-6 PUFA, could alleviate enterotoxigenic Escherichia coli (ETEC) K88-induced inflammation and injury of intestinal porcine epithelial cells 1 (IPEC-1) by modulating pyroptosis and necroptosis signalling pathways. IPEC-1 cells were cultured with or without EPA or ARA in the presence or absence of ETEC K88. EPA and ARA reduced ETEC K88 adhesion and endotoxin content in the supernatant. EPA and ARA increased transepithelial electrical resistance, decreased permeability of fluorescein isothiocyanate-labelled dextran, increased membrane protein expression of occludin, ZO-1 and claudin-1 and relieved disturbed distribution of these proteins. EPA and ARA also reduced cell necrosis ratio. EPA or ARA reduced mRNA and concentration of TNF-α, IL-6 and IL-8 and decreased mRNA abundances of intestinal toll-like receptors 4 and its downstream signals. Moreover, EPA and ARA downregulated mRNA expression of nod-like receptor protein 3 (NLRP3), caspase 1 and IL-18 and inhibited protein expression of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), gasdermin D and caspase-1. Finally, EPA and ARA reduced mRNA expression of fas-associated death domain protein, caspase 8, receptor-interacting protein kinase (RIP) 1, mixed lineage kinase-like protein (MLKL), phosphoglycerate mutase 5 (PGAM5), motility-related protein 1 (Drp1) and high mobility protein 1 (HMGB1) and inhibited protein expression of phosphorylated-RIP1, p-RIP3, p-MLKL and HMGB1. These data demonstrate that EPA and ARA prevent ETEC K88-induced cell inflammation and injury, which is partly through inhibiting pyroptosis and necroptosis signalling pathways.
Collapse
Affiliation(s)
- Kan Xiao
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yang Yang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Yang Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Qingqing Lv
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Feifei Huang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Dan Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan, 430023, People's Republic of China
| |
Collapse
|
21
|
Intestinal Epithelial Cells Modulate the Production of Enterotoxins by Porcine Enterotoxigenic E. coli Strains. Int J Mol Sci 2022; 23:ijms23126589. [PMID: 35743033 PMCID: PMC9223395 DOI: 10.3390/ijms23126589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/03/2022] [Accepted: 06/12/2022] [Indexed: 01/23/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) strains are one of the most common etiological agents of diarrhea in both human and farm animals. In addition to encoding toxins that cause diarrhea, ETEC have evolved numerous strategies to interfere with host defenses. These strategies most likely depend on the sensing of host factors, such as molecules secreted by gut epithelial cells. The present study tested whether the exposure of ETEC to factors secreted by polarized IPEC-J2 cells resulted in transcriptional changes of ETEC-derived virulence factors. Following the addition of host-derived epithelial factors, genes encoding enterotoxins, secretion-system-associated proteins, and the key regulatory molecule cyclic AMP (cAMP) receptor protein (CRP) were substantially modulated, suggesting that ETEC recognize and respond to factors produced by gut epithelial cells. To determine whether these factors were heat sensitive, the IEC-conditioned medium was incubated at 56 °C for 30 min. In most ETEC strains, heat treatment of the IEC-conditioned medium resulted in a loss of transcriptional modulation. Taken together, these data suggest that secreted epithelial factors play a role in bacterial pathogenesis by modulating the transcription of genes encoding key ETEC virulence factors. Further research is warranted to identify these secreted epithelial factors and how ETEC sense these molecules to gain a competitive advantage in the early engagement of the gut epithelium.
Collapse
|
22
|
Sheikh A, Wangdi T, Vickers TJ, Aaron B, Palmer M, Miller MJ, Kim S, Herring C, Simoes R, Crainic JA, Gildersleeve JC, van der Post S, Hansson GC, Fleckenstein JM. Enterotoxigenic Escherichia coli Degrades the Host MUC2 Mucin Barrier To Facilitate Critical Pathogen-Enterocyte Interactions in Human Small Intestine. Infect Immun 2022; 90:e0057221. [PMID: 34807735 PMCID: PMC8853678 DOI: 10.1128/iai.00572-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) isolates are genetically diverse pathological variants of E. coli defined by the production of heat-labile (LT) and/or heat-stable (ST) toxins. ETEC strains are estimated to cause hundreds of millions of cases of diarrheal illness annually. However, it is not clear that all strains are equally equipped to cause disease, and asymptomatic colonization with ETEC is common in low- to middle-income regions lacking basic sanitation and clean water where ETEC are ubiquitous. Recent molecular epidemiology studies have revealed a significant association between strains that produce EatA, a secreted autotransporter protein, and the development of symptomatic infection. Here, we demonstrate that LT stimulates production of MUC2 mucin by goblet cells in human small intestine, enhancing the protective barrier between pathogens and enterocytes. In contrast, using explants of human small intestine as well as small intestinal enteroids, we show that EatA counters this host defense by engaging and degrading the MUC2 mucin barrier to promote bacterial access to target enterocytes and ultimately toxin delivery, suggesting that EatA plays a crucial role in the molecular pathogenesis of ETEC. These findings may inform novel approaches to prevention of acute diarrheal illness as well as the sequelae associated with ETEC and other pathogens that rely on EatA and similar proteases for efficient interaction with their human hosts.
Collapse
Affiliation(s)
- Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Tamding Wangdi
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Tim J. Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Bailey Aaron
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Margot Palmer
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Mark J. Miller
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Seonyoung Kim
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Cassandra Herring
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Rita Simoes
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jennifer A. Crainic
- Center for Cancer Research, Chemical Biology Laboratory, National Cancer Institute, Fredrick, Maryland, USA
| | - Jeffrey C. Gildersleeve
- Center for Cancer Research, Chemical Biology Laboratory, National Cancer Institute, Fredrick, Maryland, USA
| | - Sjoerd van der Post
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
- Medicine Service, Veterans Affairs Medical Center, Saint Louis, Missouri, USA
| |
Collapse
|
23
|
Sabui S, Ramamoorthy K, Romero JM, Simoes RD, Fleckenstein JM, Said HM. Hypoxia inhibits colonic uptake of the microbiota-generated forms of vitamin B1 via HIF-1α-mediated transcriptional regulation of their transporters. J Biol Chem 2022; 298:101562. [PMID: 34998824 PMCID: PMC8800108 DOI: 10.1016/j.jbc.2022.101562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/29/2021] [Accepted: 01/02/2022] [Indexed: 01/19/2023] Open
Abstract
Hypoxia exerts profound effects on cell physiology, but its effect on colonic uptake of the microbiota-generated forms of vitamin B1 (i.e., thiamin pyrophosphate [TPP] and free thiamine) has not been described. Here, we used human colonic epithelial NCM460 cells and human differentiated colonoid monolayers as in vitro and ex vivo models, respectively, and were subjected to either chamber (1% O2, 5% CO2, and 94% N2) or chemically (desferrioxamine; 250 μM)-induced hypoxia followed by determination of different physiological-molecular parameters. We showed that hypoxia causes significant inhibition in TPP and free thiamin uptake by colonic NCM460 cells and colonoid monolayers; it also caused a significant reduction in the expression of TPP (SLC44A4) and free thiamin (SLC19A2 and SLC19A3) transporters and in activity of their gene promoters. Furthermore, hypoxia caused a significant induction in levels of hypoxia-inducible transcription factor (HIF)-1α but not HIF-2α. Knocking down HIF-1α using gene-specific siRNAs in NCM460 cells maintained under hypoxic conditions, on the other hand, led to a significant reversal in the inhibitory effect of hypoxia on TPP and free thiamin uptake as well as on the expression of their transporters. Finally, a marked reduction in level of expression of the nuclear factors cAMP responsive element-binding protein 1 and gut-enriched Krüppel-like factor 4 (required for activity of SLC44A4 and SLC19A2 promoters, respectively) was observed under hypoxic conditions. In summary, hypoxia causes severe inhibition in colonic TPP and free thiamin uptake that is mediated at least in part via HIF-1α-mediated transcriptional mechanisms affecting their respective transporters.
Collapse
Affiliation(s)
- Subrata Sabui
- Department of Physiology and Biophysics, UCI, Irvine, California, USA; Department of Research, VA Medical Center, Long Beach, California, USA
| | | | - Jose M Romero
- Department of Research, VA Medical Center, Long Beach, California, USA; Department of Medicine, UCI, Irvine, California, USA
| | - Rita D Simoes
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA; Department Medicine Service, Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Hamid M Said
- Department of Physiology and Biophysics, UCI, Irvine, California, USA; Department of Research, VA Medical Center, Long Beach, California, USA; Department of Medicine, UCI, Irvine, California, USA.
| |
Collapse
|
24
|
Fleckenstein JM, Sheikh A. Emerging Themes in the Molecular Pathogenesis of Enterotoxigenic Escherichia coli. J Infect Dis 2021; 224:S813-S820. [PMID: 34273153 PMCID: PMC8687053 DOI: 10.1093/infdis/jiab359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are ubiquitous diarrheal pathogens that thrive in areas lacking basic human needs of clean water and sanitation. These genetically plastic organisms cause tremendous morbidity among disadvantaged young children, in the form of both acute diarrheal illness and sequelae of malnutrition and growth impairment. The recent discovery of additional plasmid-encoded virulence factors and elucidation of their critical role in the molecular pathogenesis of ETEC may inform new approaches to the development of broadly protective vaccines. Although the pathogens have been closely linked epidemiologically with nondiarrheal sequelae, these conditions remain very poorly understood. Similarly, while canonical effects of ETEC toxins on cellular signaling promoting diarrhea are clear, emerging data suggest that these toxins may also drive changes in intestinal architecture and associated sequelae. Elucidation of molecular events underlying these changes could inform optimal approaches to vaccines that prevent acute diarrhea and ETEC-associated sequelae.
Collapse
Affiliation(s)
- James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in St Louis, School of Medicine, St Louis, Missouri, USA
- Infectious Disease Section, Medicine Service, St Louis Veterans Affairs Health Care System, St Louis, Missouri, USA
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in St Louis, School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
25
|
He F, Zhang T, Xue K, Fang Z, Jiang G, Huang S, Li K, Gu Z, Shi H, Zhang Z, Zhu H, Lin L, Li J, Xiao F, Shan H, Yan R, Li X, Yan Z. Fecal multi-omics analysis reveals diverse molecular alterations of gut ecosystem in COVID-19 patients. Anal Chim Acta 2021; 1180:338881. [PMID: 34538334 PMCID: PMC8310733 DOI: 10.1016/j.aca.2021.338881] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022]
Abstract
Gut ecosystem has profound effects on host physiology and health. Gastrointestinal (GI) symptoms were frequently observed in patients with COVID-19. Compared with other organs, gut antiviral response can result in more complicated immune responses because of the interactions between the gut microbiota and host immunity. However, there are still large knowledge gaps in the impact of COVID-19 on gut molecular profiles and commensal microbiome, hindering our comprehensive understanding of the pathogenesis of SARS-CoV-2 and the treatment of COVID-19. We performed longitudinal stool multi-omics profiling to systemically investigate the molecular phenomics alterations of gut ecosystem in COVID-19. Gut proteomes of COVID-19 were characterized by disturbed immune, proteolysis and redox homeostasis. The expression and glycosylation of proteins involved in neutrophil degranulation and migration were suppressed, while those of proteases were upregulated. The variable domains of Ig heavy chains were downregulated and the overall glycosylation of IgA heavy chain constant regions, IgGFc-binding protein, and J chain were suppressed with glycan-specific variations. There was a reduction of beneficial gut bacteria and an enrichment of bacteria derived deleterious metabolites potentially associated with multiple types of diseases (such as ethyl glucuronide). The reduction of Ig heave chain variable domains may contribute to the increase of some Bacteroidetes species. Many bacteria ceramide lipids with a C17-sphingoid based were downregulated in COVID-19. In many cases, the gut phenome did not restore two months after symptom onset. Our study indicates widely disturbed gut molecular profiles which may play a role in the development of symptoms in COVID-19. Our findings also emphasis the need for ongoing investigation of the long-term gut molecular and microbial alterations during COVID-19 recovery process. Considering the gut ecosystem as a potential target could offer a valuable approach in managing the disease.
Collapse
Affiliation(s)
- Feixiang He
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China,Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Ting Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Kewen Xue
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Zhaoxiong Fang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Siwen Huang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Kexue Li
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Zhiqiang Gu
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Honggang Shi
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Zhenyi Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Huijin Zhu
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Lu Lin
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Jialin Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Fei Xiao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China,Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao,Corresponding author
| | - Xiaofeng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China,Corresponding author
| | - Zhixiang Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China,Corresponding author
| |
Collapse
|
26
|
Ramamoorthy K, Sabui S, Srinivasan P, Al-Juburi S, Pham Q, Chu BD, Simoes RD, Fleckenstein JM, Said HM. Effect of chronic alcohol exposure on gut vitamin B7 uptake: involvement of epigenetic mechanisms and effect of alcohol metabolites. Am J Physiol Gastrointest Liver Physiol 2021; 321:G123-G133. [PMID: 34077272 PMCID: PMC8410103 DOI: 10.1152/ajpgi.00144.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Vitamin B7 (biotin) is essential for normal health and its deficiency/suboptimal levels occur in a variety of conditions including chronic alcoholism. Mammals, including humans, obtain biotin from diet and gut-microbiota via absorption along the intestinal tract. The absorption process is carrier mediated and involves the sodium-dependent multivitamin transporter (SMVT; SLC5A6). We have previously shown that chronic alcohol exposure significantly inhibits intestinal/colonic biotin uptake via suppression of Slc5a6 transcription in animal and cell line models. However, little is known about the transcriptional/epigenetic factors that mediate this suppression. In addition, the effect of alcohol metabolites (generated via alcohol metabolism by gut microbiota and host tissues) on biotin uptake is still unknown. To address these questions, we first demonstrated that chronic alcohol exposure inhibits small intestinal and colonic biotin uptake and SMVT expression in human differentiated enteroid and colonoid monolayers. We then showed that chronic alcohol exposures of both, Caco-2 cells and mice, are associated with a significant suppression in expression of the nuclear factor KLF-4 (needed for Slc5a6 promoter activity), as well as with epigenetic alterations (histone modifications). We also found that chronic exposure of NCM460 human colonic epithelial cells as well as human differentiated colonoid monolayers, to alcohol metabolites (acetaldehyde, ethyl palmitate, ethyl oleate) significantly inhibited biotin uptake and SMVT expression. These findings shed light onto the molecular/epigenetic mechanisms that mediate the inhibitory effect of chronic alcohol exposure on intestinal biotin uptake. They further show that alcohol metabolites are also capable of inhibiting biotin uptake in the gut.NEW & NOTEWORTHY Using complementary models, including human differentiated enteroid and colonoid monolayers, this study shows the involvement of molecular and epigenetic mechanisms in mediating the inhibitory effect of chronic alcohol exposure on biotin uptake along the intestinal tract. The study also shows that alcohol metabolites (generated by gut microbiota and host tissues) cause inhibition in gut biotin uptake.
Collapse
Affiliation(s)
- Kalidas Ramamoorthy
- 1Department of Physiology/Biophysics, University of California, Irvine, California
| | - Subrata Sabui
- 1Department of Physiology/Biophysics, University of California, Irvine, California,5Veterans Affairs Medical Center, Long Beach, California
| | - Padmanabhan Srinivasan
- 1Department of Physiology/Biophysics, University of California, Irvine, California,5Veterans Affairs Medical Center, Long Beach, California
| | - Saleh Al-Juburi
- 1Department of Physiology/Biophysics, University of California, Irvine, California
| | - Quang Pham
- 1Department of Physiology/Biophysics, University of California, Irvine, California
| | - Brian D. Chu
- 1Department of Physiology/Biophysics, University of California, Irvine, California,5Veterans Affairs Medical Center, Long Beach, California
| | - Rita D. Simoes
- 3Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - James M. Fleckenstein
- 3Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri,4Veterans Affairs Medical Center, St. Louis Missouri
| | - Hamid M. Said
- 1Department of Physiology/Biophysics, University of California, Irvine, California,2Department of Medicine, University of California, Irvine, California,5Veterans Affairs Medical Center, Long Beach, California
| |
Collapse
|
27
|
Khalil I, Walker R, Porter CK, Muhib F, Chilengi R, Cravioto A, Guerrant R, Svennerholm AM, Qadri F, Baqar S, Kosek M, Kang G, Lanata C, Armah G, Wierzba T, Hasso-Agopsowicz M, Giersing B, Louis Bourgeois A. Enterotoxigenic Escherichia coli (ETEC) vaccines: Priority activities to enable product development, licensure, and global access. Vaccine 2021; 39:4266-4277. [PMID: 33965254 PMCID: PMC8273896 DOI: 10.1016/j.vaccine.2021.04.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023]
Abstract
Diarrhoeal disease attributable to enterotoxigenic Escherichia coli (ETEC) causes substantial morbidity and mortality predominantly in paediatric populations in low- and middle-income countries. In addition to acute illness, there is an increasing appreciation of the long-term consequences of enteric infections, including ETEC, on childhood growth and development. Provision of potable water and sanitation and appropriate clinical care for acute illness are critical to reduce the ETEC burden. However, these interventions are not always practical and may not achieve equitable and sustainable coverage. Vaccination may be the most cost-effective and equitable means of primary prevention; however, additional data are needed to accelerate the investment and guide the decision-making process for ETEC vaccines. First, to understand and quantify the ETEC disease burden, additional data are needed on the association between ETEC infection and physical and cognitive stunting as well as delayed educational attainment. Furthermore, the role of inappropriate or inadequate antibiotic treatment of ETEC-attributable diarrhoea may contribute to the development of antimicrobial resistance (AMR) and needs further elucidation. An ETEC vaccine that mitigates acute diarrhoeal illness and minimizes the longer-term disease manifestations could have significant public health impact and be a cost-effective countermeasure. Herein we review the ETEC vaccine pipeline, led by candidates compatible with the general parameters of the Preferred Product Characteristics (PPC) recently developed by the World Health Organization. Additionally, we have developed an ETEC Vaccine Development Strategy to provide a framework to underpin priority activities for researchers, funders and vaccine manufacturers, with the goal of addressing globally unmet data needs in the areas of research, product development, and policy, as well as commercialization and delivery. The strategy also aims to guide prioritization and co-ordination of the priority activities needed to minimize the timeline to licensure and use of ETEC vaccines, especially in in low- and middle-income countries, where they are most urgently needed.
Collapse
Affiliation(s)
| | | | | | | | - Roma Chilengi
- Centre for Infectious Disease Research in Zambia, Zambia
| | | | | | | | | | - Shahida Baqar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA
| | | | | | | | - George Armah
- Noguchi Memorial Institute for Medical Research, Ghana
| | | | | | | | | |
Collapse
|
28
|
Walker R, Kaminski RW, Porter C, Choy RKM, White JA, Fleckenstein JM, Cassels F, Bourgeois L. Vaccines for Protecting Infants from Bacterial Causes of Diarrheal Disease. Microorganisms 2021; 9:1382. [PMID: 34202102 PMCID: PMC8303436 DOI: 10.3390/microorganisms9071382] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
The global diarrheal disease burden for Shigella, enterotoxigenic Escherichia coli (ETEC), and Campylobacter is estimated to be 88M, 75M, and 75M cases annually, respectively. A vaccine against this target trio of enteric pathogens could address about one-third of diarrhea cases in children. All three of these pathogens contribute to growth stunting and have demonstrated increasing resistance to antimicrobial agents. Several combinations of antigens are now recognized that could be effective for inducing protective immunity against each of the three target pathogens in a single vaccine for oral administration or parenteral injection. The vaccine combinations proposed here would result in a final product consistent with the World Health Organization's (WHO) preferred product characteristics for ETEC and Shigella vaccines, and improve the vaccine prospects for support from Gavi, the Vaccine Alliance, and widespread uptake by low- and middle-income countries' (LMIC) public health stakeholders. Broadly protective antigens will enable multi-pathogen vaccines to be efficiently developed and cost-effective. This review describes how emerging discoveries for each pathogen component of the target trio could be used to make vaccines, which could help reduce a major cause of poor health, reduced cognitive development, lost economic productivity, and poverty in many parts of the world.
Collapse
Affiliation(s)
- Richard Walker
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA;
| | - Robert W. Kaminski
- Department of Diarrheal Disease Research, Walter Reed Institute of Research, Silver Spring, MD 20910, USA;
| | - Chad Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910, USA;
| | - Robert K. M. Choy
- Center for Vaccine Innovation and Access, PATH, San Francisco, CA 94108, USA;
| | - Jessica A. White
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA; (J.A.W.); (F.C.)
| | - James M. Fleckenstein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Medicine Service, Saint Louis VA Health Care System, St. Louis, MO 63106, USA
| | - Fred Cassels
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA; (J.A.W.); (F.C.)
| | - Louis Bourgeois
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA;
| |
Collapse
|