1
|
Maeda K, Zachos NC, Orzalli MH, Schmieder SS, Chang D, Bugda Gwilt K, Doucet M, Baetz NW, Lee S, Crawford SE, Estes MK, Kagan JC, Turner JR, Lencer WI. Depletion of the apical endosome in response to viruses and bacterial toxins provides cell-autonomous host defense at mucosal surfaces. Cell Host Microbe 2022; 30:216-231.e5. [PMID: 35143768 PMCID: PMC8852832 DOI: 10.1016/j.chom.2021.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 10/28/2021] [Accepted: 12/17/2021] [Indexed: 11/22/2022]
Abstract
Polarized epithelial cells form an essential barrier against infection at mucosal surfaces. Many pathogens breach this barrier to cause disease, often by co-opting cellular endocytosis mechanisms to enter the cell through the lumenal (apical) cell surface. We recently discovered that the loss of the cell polarity gene PARD6B selectively diminishes apical endosome function. Here, we find that in response to the entry of certain viruses and bacterial toxins into the epithelial cells via the apical membrane, PARD6B and aPKC, two components of the PARD6B-aPKC-Cdc42 apical polarity complex, undergo rapid proteasome-dependent degradation. The perturbation of apical membrane glycosphingolipids by toxin- or virus-binding initiates degradation of PARD6B. The loss of PARD6B causes the depletion of apical endosome function and renders the cell resistant to further infection from the lumenal cell surface, thus enabling a form of cell-autonomous host defense.
Collapse
Affiliation(s)
- Keiko Maeda
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas C Zachos
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Megan H Orzalli
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stefanie S Schmieder
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Denis Chang
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katlynn Bugda Gwilt
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michele Doucet
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas W Baetz
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sun Lee
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sue E Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, MS: BCM-385, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jonathan C Kagan
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jerrold R Turner
- Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | - Wayne I Lencer
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Melnik LI, Garry RF. Enterotoxigenic Escherichia coli Heat-Stable Toxin and Ebola Virus Delta Peptide: Similarities and Differences. Pathogens 2022; 11:pathogens11020170. [PMID: 35215114 PMCID: PMC8878840 DOI: 10.3390/pathogens11020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) STb toxin exhibits striking structural similarity to Ebola virus (EBOV) delta peptide. Both ETEC and EBOV delta peptide are enterotoxins. Comparison of the structural and functional similarities and differences of these two toxins illuminates features that are important in induction of pathogenesis by a bacterial and viral pathogen.
Collapse
Affiliation(s)
- Lilia I. Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Viral Hemorrhagic Fever Consortium, New Orleans, LA 70112, USA
- Correspondence: ; Tel.: +1-(504)988-3818
| | - Robert F. Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Viral Hemorrhagic Fever Consortium, New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Melnik LI, Guha S, Ghimire J, Smither AR, Beddingfield BJ, Hoffmann AR, Sun L, Ungerleider NA, Baddoo MC, Flemington EK, Gallaher WR, Wimley WC, Garry RF. Ebola virus delta peptide is an enterotoxin. Cell Rep 2022; 38:110172. [PMID: 34986351 DOI: 10.1016/j.celrep.2021.110172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/27/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
During the 2013-2016 West African (WA) Ebola virus (EBOV) outbreak, severe gastrointestinal symptoms were common in patients and associated with poor outcome. Delta peptide is a conserved product of post-translational processing of the abundant EBOV soluble glycoprotein (sGP). The murine ligated ileal loop model was used to demonstrate that delta peptide is a potent enterotoxin. Dramatic intestinal fluid accumulation follows injection of biologically relevant amounts of delta peptide into ileal loops, along with gross alteration of villous architecture and loss of goblet cells. Transcriptomic analyses show that delta peptide triggers damage response and cell survival pathways and downregulates expression of transporters and exchangers. Induction of diarrhea by delta peptide occurs via cellular damage and regulation of genes that encode proteins involved in fluid secretion. While distinct differences exist between the ileal loop murine model and EBOV infection in humans, these results suggest that delta peptide may contribute to EBOV-induced gastrointestinal pathology.
Collapse
Affiliation(s)
- Lilia I Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shantanu Guha
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jenisha Ghimire
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Allison R Smither
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Brandon J Beddingfield
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Andrew R Hoffmann
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Leisheng Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | - Melody C Baddoo
- Tulane Cancer Center, Tulane University, New Orleans, LA 70112, USA
| | | | - William R Gallaher
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA 70112, USA; Mockingbird Nature Research Group, Pearl River, LA 70452, USA
| | - William C Wimley
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Zalgen Labs, Germantown, MD 20876, USA.
| |
Collapse
|
4
|
Koepsell H. Glucose transporters in the small intestine in health and disease. Pflugers Arch 2020; 472:1207-1248. [PMID: 32829466 PMCID: PMC7462918 DOI: 10.1007/s00424-020-02439-5] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 12/23/2022]
Abstract
Absorption of monosaccharides is mainly mediated by Na+-D-glucose cotransporter SGLT1 and the facititative transporters GLUT2 and GLUT5. SGLT1 and GLUT2 are relevant for absorption of D-glucose and D-galactose while GLUT5 is relevant for D-fructose absorption. SGLT1 and GLUT5 are constantly localized in the brush border membrane (BBM) of enterocytes, whereas GLUT2 is localized in the basolateral membrane (BLM) or the BBM plus BLM at low and high luminal D-glucose concentrations, respectively. At high luminal D-glucose, the abundance SGLT1 in the BBM is increased. Hence, D-glucose absorption at low luminal glucose is mediated via SGLT1 in the BBM and GLUT2 in the BLM whereas high-capacity D-glucose absorption at high luminal glucose is mediated by SGLT1 plus GLUT2 in the BBM and GLUT2 in the BLM. The review describes functions and regulations of SGLT1, GLUT2, and GLUT5 in the small intestine including diurnal variations and carbohydrate-dependent regulations. Also, the roles of SGLT1 and GLUT2 for secretion of enterohormones are discussed. Furthermore, diseases are described that are caused by malfunctions of small intestinal monosaccharide transporters, such as glucose-galactose malabsorption, Fanconi syndrome, and fructose intolerance. Moreover, it is reported how diabetes, small intestinal inflammation, parental nutrition, bariatric surgery, and metformin treatment affect expression of monosaccharide transporters in the small intestine. Finally, food components that decrease D-glucose absorption and drugs in development that inhibit or downregulate SGLT1 in the small intestine are compiled. Models for regulations and combined functions of glucose transporters, and for interplay between D-fructose transport and metabolism, are discussed.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstr 6, 97070, Würzburg, Germany.
| |
Collapse
|
5
|
Cruz LF, Figueiredo GFD, Pedro LP, Amorin YM, Andrade JT, Passos TF, Rodrigues FF, Souza ILA, Gonçalves TPR, Dos Santos Lima LAR, Ferreira JMS, Araújo MGDF. Umbelliferone (7-hydroxycoumarin): A non-toxic antidiarrheal and antiulcerogenic coumarin. Biomed Pharmacother 2020; 129:110432. [PMID: 32768935 DOI: 10.1016/j.biopha.2020.110432] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/09/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal diseases are very common problems; available treatments are very limited and come with a range of side effects. Coumarins are an extensive class of phenolic compounds that can be found in plants, fungi and bacteria. The 7-hydroxycoumarin, also known as umbelliferone (UMB), is a compound that comes from coumarin and has been showing biological activities in other studies. As of this scenario, the present study was designed to evaluate the acute oral toxicity, mutagenic, antidiarrheal, anti-bacterial, and antiulcerogenic effects, and antioxidant capacity of UMB. An investigation was conducted through the hippocratic screening method and through histopathological analysis in animals to evaluate the effects of acute oral administration of a dose of 50, 100 and 200 mg/kg of UMB. A micronucleus test on peripheral blood of Swiss mice, which were orally treated with three doses (50, 100 and 200 mg/kg), was conducted to evaluate mutagenic activities. The antiulcerogenic activity was accomplished through the ethanol-induced damage method. Antidiarrheal activities were tested for inducing diarrhea with castor oil and evaluating intestinal transit duration; additionally, the antimicrobial effect against some enteropathogenic bacteria was analyzed. Finally, the antioxidant capability was determined by the capacity of the UMB sample to kidnap the stable radical 2,2-diphenyl-1-picrylhydrazyl. Of the evaluated doses, signs of toxicity after acute administration of the compound were not observed. UMB presented antiulcerogenic activity (100 and 200 mg/kg), which was explained because of its antioxidant capacity. A gastro protective effect was similar to the positive control, and the UMB was able to significantly reduce intestinal transit, and also diarrheal symptoms. Furthermore, UMB had an anti-bacterial effect with minimum inhibitory concentration fluctuating between 62.5 and 1000 μg/mL. Based on these findings, we can suggest that UMB has important biological activities in vivo and in vitro and is not toxic under the evaluated circumstances, which demonstrates its large potential for pharmacological use.
Collapse
Affiliation(s)
- Luisa Ferreira Cruz
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil.
| | - Geisa Fantini de Figueiredo
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Lidiane Paula Pedro
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Yuri Martins Amorin
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Jessica Tauany Andrade
- Laboratory of Medical Microbiology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Tiago Faustino Passos
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Felipe Fernandes Rodrigues
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Israel Lucas Antunes Souza
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Thaís Paula Rodrigues Gonçalves
- Laboratory of Phytochemistry, Federal University of São João Del-Rei, Campus Centro Oeste Dona Lindu, Divinópolis, MG, Brazil
| | | | - Jaqueline Maria Siqueira Ferreira
- Laboratory of Medical Microbiology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| | - Marcelo Gonzaga de Freitas Araújo
- Laboratory of Pharmacology, Federal University of São João Del-Rei, Sebastião Gonçalves Coelho, 400, Chanadour, 35501-296, Divinópolis, MG, Brazil
| |
Collapse
|
6
|
Huang H, Pu Y, Liao D, Zhu Z, Wang J, Cui Y. The expression of calcium-sensing receptor during rotavirus induced diarrhea in neonatal mice. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2019. [DOI: 10.23736/s0393-3660.18.03910-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
7
|
McCowan C, Crameri S, Kocak A, Shan S, Fegan M, Forshaw D, Rubbenstroth D, Chen H, Holmes C, Harper J, Dearnley M, Batovska J, Bergfeld J, Walker C, Wang J. A novel group A rotavirus associated with acute illness and hepatic necrosis in pigeons (Columba livia), in Australia. PLoS One 2018; 13:e0203853. [PMID: 30204797 PMCID: PMC6133385 DOI: 10.1371/journal.pone.0203853] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
Cases of vomiting and diarrhoea were reported in racing pigeons in Western Australia in May, 2016. Morbidity and mortality rates were high. Similar clinical disease was seen in Victoria in December and by early 2017 had been reported in all states except the Northern Territory, in different classes of domestic pigeon–racing, fancy and meat bird–and in a flock of feral pigeons. Autopsy findings were frequently unremarkable; histological examination demonstrated significant hepatic necrosis as the major and consistent lesion, often with minimal inflammatory infiltration. Negative contrast tissue suspension and thin section transmission electron microscopy of liver demonstrated virus particles consistent with a member of the Reoviridae. Inoculation of trypsin-treated Vero, MDBK and MA-104 cell lines resulted in cytopathic changes at two days after infection. Next generation sequencing was undertaken using fresh liver samples and a previously undescribed group A rotavirus (genotype G18P[17]) of avian origin was identified and the virus was isolated in several cell lines. A q-RT-PCR assay was developed and used to screen a wider range of samples, including recovered birds. Episodes of disease have continued to occur and to reoccur in previously recovered lofts, with variable virulence reported. This is the first report of a rotavirus associated with hepatic necrosis in any avian species.
Collapse
Affiliation(s)
| | - Sandra Crameri
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Ayfer Kocak
- Agriculture Victoria, Bundoora, Victoria, Australia
| | - Songhua Shan
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Mark Fegan
- Agriculture Victoria, Bundoora, Victoria, Australia
| | - David Forshaw
- Department of Primary Industries and Regional Development, Albany, Western Australia, Australia
| | - Dennis Rubbenstroth
- Institute of Virology, Medical Center–University of Freiburg, Freiburg, Germany
- Institute for Diagnostic Virology, Friedrich-Loeffler-Institute (FLI), Greifswald–Insel Riems, Germany
| | - Honglei Chen
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Clare Holmes
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Jenni Harper
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Megan Dearnley
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Jana Batovska
- Agriculture Victoria, Bundoora, Victoria, Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, Victoria, Australia
| | - Jemma Bergfeld
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Colin Walker
- Melbourne Bird Veterinary Clinic, Scoresby, Melbourne, Australia
| | - Jianning Wang
- Australian Animal Health Laboratory, Geelong, Victoria, Australia
| |
Collapse
|
8
|
Urdaci MC, Lefevre M, Lafforgue G, Cartier C, Rodriguez B, Fioramonti J. Antidiarrheal Action of Bacillus subtilis CU1 CNCM I-2745 and Lactobacillus plantarum CNCM I-4547 in Mice. Front Microbiol 2018; 9:1537. [PMID: 30042756 PMCID: PMC6048234 DOI: 10.3389/fmicb.2018.01537] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/20/2018] [Indexed: 12/29/2022] Open
Abstract
Preventive actions of probiotics as antidiarrheal agents are well documented, but their mechanisms are poorly understood. Two selected probiotics, Bacillus subtilis CU1 and Lactobacillus plantarum CNCM I-4547, were tested in mouse experimental models of diarrhea and the possible mechanisms of action were investigated. Diarrhea was induced in mice by oral castor oil administration or by i.v. injection of lipopolysaccharide (LPS) of Salmonella enteritis. The antidiarrheal drug loperamide was used as control. Fecal water excretion was quantified for 2 h and paracellular permeability and electrical parameters of the colon were assessed in Ussing chambers. The expression of colonic exchangers or channels and of Toll-like receptor 4 (TLR4) was assessed by immunohistochemistry. Prophylactic treatment with B. subtilis CU1 or with L. plantarum CNCM I-4547 reduced LPS-induced diarrhea. The reduction of water excretion was in the same range as those induced by loperamide. In the castor oil model, this effect was only observed with B. subtilis CU1. The two probiotic treatments abolished the increase in paracellular permeability induced by LPS, but not by castor oil. However, only L. plantarum CNCM I-4547 treatment decreased the colonic expression of TLR-4. After B. subtilis CU1, colonic expression of cystic fibrosis transmembrane conductance regulator (CFTR) was reduced and that of Na+/H+ exchanger 3 (NHE3) increased. B. subtilis CU1 may increase the capacity of the colon to absorb excess of water in diarrheic conditions by acting on CFTR and NHE3 expression. The two probiotics strains showed an impact on diarrhea through limitation of water excretion that may involve paracellular permeability or electrolyte transport for L. plantarum CNCM I-4547 and B. subtilis CU1 respectively.
Collapse
Affiliation(s)
- Maria C Urdaci
- Microbiology Laboratory, UMR 5248, Bordeaux Sciences Agro, University of Bordeaux, Gradignan, France
| | - Marie Lefevre
- Lesaffre Human Care, Lesaffre Group, Marcq-en-Baroeul, France
| | - Guylene Lafforgue
- Neuro-Gastroenterology and Nutrition Unit, INRA, Toulouse, France.,Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Christel Cartier
- Neuro-Gastroenterology and Nutrition Unit, INRA, Toulouse, France.,Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Jean Fioramonti
- Neuro-Gastroenterology and Nutrition Unit, INRA, Toulouse, France.,Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
9
|
Das S, Jayaratne R, Barrett KE. The Role of Ion Transporters in the Pathophysiology of Infectious Diarrhea. Cell Mol Gastroenterol Hepatol 2018; 6:33-45. [PMID: 29928670 PMCID: PMC6007821 DOI: 10.1016/j.jcmgh.2018.02.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022]
Abstract
Every year, enteric infections and associated diarrhea kill millions of people. The situation is compounded by increases in the number of enteric pathogens that are acquiring resistance to antibiotics, as well as (hitherto) a relative paucity of information on host molecular targets that may contribute to diarrhea. Many forms of diarrheal disease depend on the dysregulation of intestinal ion transporters, and an associated imbalance between secretory and absorptive functions of the intestinal epithelium. A number of major transporters have been implicated in the pathogenesis of diarrheal diseases and thus an understanding of their expression, localization, and regulation after infection with various bacteria, viruses, and protozoa likely will prove critical in designing new therapies. This article surveys our understanding of transporters that are modulated by specific pathogens and the mechanism(s) involved, thereby illuminating targets that might be exploited for new therapeutic approaches.
Collapse
Key Words
- ATP, adenosine triphosphate
- ATPase, adenosine triphosphatase
- CDI, Clostridium difficile infection
- CFTR, cystic fibrosis transmembrane conductance regulator
- CLCA1, chloride channel accessory 1
- CT, cholera toxin
- CXCR2, C-X-C motif chemokine receptor 2
- DRA, down-regulated in adenoma
- Diarrhea
- ENaC, epithelial sodium channel
- EPEC, enteropathogenic Escherichia coli
- ETEC, enterotoxigenic Escherichia coli
- Enteric Pathogen
- Epithelium
- EspG, Escherichia coli secreted protein G
- GPR39, G-protein coupled receptor 39
- Ion Transport
- KCC, potassium-chloride cotransporter
- LPA, lysophosphatidic acid
- LT, heat-labile toxin
- NHE, sodium/hydrogen exchanger
- NHERF2, sodium/hydrogen exchanger regulatory factor 2
- NKCC, sodium-potassium-2 chloride cotransporter
- ORT, oral rehydration therapy
- PKC, protein kinase C
- SGLT1, sodium-glucose cotransporter 1
- SLC, solute carrier
- ST, heat-stabile toxin
- TNF, tumor necrosis factor
- Tcd, Clostridium difficile toxin
- ZnR, zinc sensing receptor
- cAMP, adenosine 3′,5′-cyclic monophosphate
Collapse
Affiliation(s)
- Soumita Das
- Department of Pathology, University of California San Diego School of Medicine, La Jolla, California
| | - Rashini Jayaratne
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California
| | - Kim E. Barrett
- Department of Medicine, University of California San Diego School of Medicine, La Jolla, California,Correspondence Address correspondence to: Kim E. Barrett, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0063. fax: (858) 246-1788.
| |
Collapse
|
10
|
|
11
|
Sauvanet C, Wayt J, Pelaseyed T, Bretscher A. Structure, Regulation, and Functional Diversity of Microvilli on the Apical Domain of Epithelial Cells. Annu Rev Cell Dev Biol 2015; 31:593-621. [DOI: 10.1146/annurev-cellbio-100814-125234] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Cécile Sauvanet
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Jessica Wayt
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Thaher Pelaseyed
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| | - Anthony Bretscher
- Department of Molecular Biology and Genetics and Weill Institute for Molecular and Cell Biology, Cornell University, Ithaca, New York 14853;
| |
Collapse
|
12
|
Abstract
Diarrhoeal disease remains a major health burden worldwide. Secretory diarrhoeas are caused by certain bacterial and viral infections, inflammatory processes, drugs and genetic disorders. Fluid secretion across the intestinal epithelium in secretory diarrhoeas involves multiple ion and solute transporters, as well as activation of cyclic nucleotide and Ca(2+) signalling pathways. In many secretory diarrhoeas, activation of Cl(-) channels in the apical membrane of enterocytes, including the cystic fibrosis transmembrane conductance regulator and Ca(2+)-activated Cl(-) channels, increases fluid secretion, while inhibition of Na(+) transport reduces fluid absorption. Current treatment of diarrhoea includes replacement of fluid and electrolyte losses using oral rehydration solutions, and drugs targeting intestinal motility or fluid secretion. Therapeutics in the development pipeline target intestinal ion channels and transporters, regulatory proteins and cell surface receptors. This Review describes pathogenic mechanisms of secretory diarrhoea, current and emerging therapeutics, and the challenges in developing antidiarrhoeal therapeutics.
Collapse
Affiliation(s)
- Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Mark Donowitz
- Departments of Physiology and Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Ross 925, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, 1246 Health Sciences East Tower, University of California, 500 Parnassus Avenue, San Francisco, CA 94143, USA
| |
Collapse
|
13
|
Scott C, Griffin S. Viroporins: structure, function and potential as antiviral targets. J Gen Virol 2015; 96:2000-2027. [PMID: 26023149 DOI: 10.1099/vir.0.000201] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The channel-forming activity of a family of small, hydrophobic integral membrane proteins termed 'viroporins' is essential to the life cycles of an increasingly diverse range of RNA and DNA viruses, generating significant interest in targeting these proteins for antiviral development. Viroporins vary greatly in terms of their atomic structure and can perform multiple functions during the virus life cycle, including those distinct from their role as oligomeric membrane channels. Recent progress has seen an explosion in both the identification and understanding of many such proteins encoded by highly significant pathogens, yet the prototypic M2 proton channel of influenza A virus remains the only example of a viroporin with provenance as an antiviral drug target. This review attempts to summarize our current understanding of the channel-forming functions for key members of this growing family, including recent progress in structural studies and drug discovery research, as well as novel insights into the life cycles of many viruses revealed by a requirement for viroporin activity. Ultimately, given the successes of drugs targeting ion channels in other areas of medicine, unlocking the therapeutic potential of viroporins represents a valuable goal for many of the most significant viral challenges to human and animal health.
Collapse
Affiliation(s)
- Claire Scott
- Leeds Institute of Cancer & Pathology and Leeds CRUK Clinical Centre, Faculty of Medicine and Health, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| | - Stephen Griffin
- Leeds Institute of Cancer & Pathology and Leeds CRUK Clinical Centre, Faculty of Medicine and Health, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| |
Collapse
|
14
|
Yakshe KA, Franklin ZD, Ball JM. Rotaviruses: Extraction and Isolation of RNA, Reassortant Strains, and NSP4 Protein. ACTA ACUST UNITED AC 2015; 37:15C.6.1-44. [PMID: 26344218 DOI: 10.1002/9780471729259.mc15c06s37] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Rotavirus (RV) contains 11 double-stranded RNA segments that encode for twelve structural and nonstructural proteins. The separation and isolation of viral RNA is a necessary precursor for many experimental techniques and can be useful for rapid RV RNA typing and sequencing of different rotavirus strains. The segmented genome enables RV to recombine easily. These recombinant viruses are essential for many purposes, including generation of potential vaccine strains. Rotavirus gene 10 expresses the viral enterotoxin, NSP4, which has been the focus of several studies due to the influence of NSP4 on rotavirus replication, morphogenesis, and pathogenesis. This unit will describe the isolation and separation of viral RNAs, the production characterization of recombinant RV in culture, and the expression and isolation of NSP4 in mammalian and insect cells.
Collapse
Affiliation(s)
- Krystle A Yakshe
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Zachary D Franklin
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - Judith M Ball
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
15
|
Huang H, Liao D, Liang L, Song L, Zhao W. Genistein inhibits rotavirus replication and upregulates AQP4 expression in rotavirus-infected Caco-2 cells. Arch Virol 2015; 160:1421-33. [PMID: 25877820 DOI: 10.1007/s00705-015-2404-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/19/2015] [Indexed: 11/29/2022]
Abstract
Rotavirus (RV) is the primary cause of severe dehydrating gastroenteritis and acute diarrheal disease in infants and young children. Previous studies have revealed that genistein can inhibit the infectivity of enveloped or nonenveloped viruses. Although the biological properties of genistein are well studied, the mechanisms of action underlying their anti-rotavirus properties have not been fully elucidated. Here, we report that genistein significantly inhibits RV-Wa replication in vitro by repressing viral RNA transcripts, and possibly viral protein synthesis. Interestingly, we also found that aquaporin 4 (AQP4) mRNA and protein expression, which was downregulated in RV-infected Caco-2 cells, can be upregulated by genistein in a time- and dose-dependent manner. Further experiments confirmed that genistein triggers CREB phosphorylation through PKA activation and subsequently promotes AQP4 gene transcription. These findings suggest that the pathophysiological mechanism of RV infection involves decreased expression of AQP4 and that genistein may be a useful candidate for developing a new anti-RV strategy by inhibiting rotavirus replication and upregulating AQP4 expression via the cAMP/PKA/CREB signaling pathway. Further studies on the effect of genistein on RV-induced diarrhea are warranted.
Collapse
Affiliation(s)
- Haohai Huang
- School of Pharmacy, Guangdong Medical College, No. 1, Xincheng Road of Songshan Lake Science and Technology Industry Park, Dongguan, 523808, Guangdong, China
| | | | | | | | | |
Collapse
|
16
|
Dormitzer PR. Rotaviruses. MANDELL, DOUGLAS, AND BENNETT'S PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES 2015:1854-1864.e4. [DOI: 10.1016/b978-1-4557-4801-3.00152-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
17
|
Notari L, Riera DC, Sun R, Bohl JA, McLean LP, Madden KB, van Rooijen N, Vanuytsel T, Urban JF, Zhao A, Shea-Donohue T. Role of macrophages in the altered epithelial function during a type 2 immune response induced by enteric nematode infection. PLoS One 2014; 9:e84763. [PMID: 24465430 PMCID: PMC3900397 DOI: 10.1371/journal.pone.0084763] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 11/18/2013] [Indexed: 01/10/2023] Open
Abstract
Parasitic enteric nematodes induce a type 2 immune response characterized by increased production of Th2 cytokines, IL-4 and IL-13, and recruitment of alternatively activated macrophages (M2) to the site of infection. Nematode infection is associated with changes in epithelial permeability and inhibition of sodium-linked glucose absorption, but the role of M2 in these effects is unknown. Clodronate-containing liposomes were administered prior to and during nematode infection to deplete macrophages and prevent the development of M2 in response to infection with Nippostrongylus brasiliensis. The inhibition of epithelial glucose absorption that is associated with nematode infection involved a macrophage-dependent reduction in SGLT1 activity, with no change in receptor expression, and a macrophage-independent down-regulation of GLUT2 expression. The reduced transport of glucose into the enterocyte is compensated partially by an up-regulation of the constitutive GLUT1 transporter consistent with stress-induced activation of HIF-1α. Thus, nematode infection results in a “lean” epithelial phenotype that features decreased SGLT1 activity, decreased expression of GLUT2 and an emergent dependence on GLUT1 for glucose uptake into the enterocyte. Macrophages do not play a role in enteric nematode infection-induced changes in epithelial barrier function. There is a greater contribution, however, of paracellular absorption of glucose to supply the energy demands of host resistance. These data provide further evidence of the ability of macrophages to alter glucose metabolism of neighboring cells.
Collapse
Affiliation(s)
- Luigi Notari
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Diana C. Riera
- Department of Pediatrics, Walter Reed Army Medical Center, Washington, DC, United States of America
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Rex Sun
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer A. Bohl
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Leon P. McLean
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Kathleen B. Madden
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Nico van Rooijen
- Vrije Universiteit, VUMC, Department of Molecular Cell Biology, Amsterdam, The Netherlands
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders, University Hospital Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Joseph F. Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, & Immunology Laboratory, Beltsville, Maryland, United States of America
| | - Aiping Zhao
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Terez Shea-Donohue
- Department of Medicine and Mucosal Biology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Lactase persistence and augmented salivary alpha-amylase gene copy numbers might have been selected by the combined toxic effects of gluten and (food born) pathogens. Med Hypotheses 2014; 82:326-34. [PMID: 24472865 DOI: 10.1016/j.mehy.2013.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 12/02/2013] [Accepted: 12/24/2013] [Indexed: 12/26/2022]
Abstract
Various positively selected adaptations to new nutrients have been identified. Lactase persistence is among the best known, conferring the ability for drinking milk at post weaning age. An augmented number of amylase gene (AMY1) copies, giving rise to higher salivary amylase activity, has been implicated in the consumption of starch-rich foods. Higher AMY1 copy numbers have been demonstrated in populations with recent histories of starchy-rich diets. It is however questionable whether the resulting polymorphisms have exerted positive selection only by providing easily available sources of macro and micronutrients. Humans have explored new environments more than any other animal. Novel environments challenge the host, but especially its immune system with new climatic conditions, food and especially pathogens. With the advent of the agricultural revolution and the concurrent domestication of cattle came new pathogens. We contend that specific new food ingredients (e.g., gluten) and novel pathogens drove selection for lactase persistence and higher AMY gene copy numbers. Both adaptations provide ample glucose for activating the sodium glucose-dependent co-transporter 1 (SGLT1), which is the principal glucose, sodium and water transporter in the gastro-intestinal tract. Their rapid uptake confers protection against potentially lethal dehydration, hyponatremia and ultimately multiple organ failure. Oral rehydration therapy aims at SGLT1 activity and is the current treatment of choice for chronic diarrhoea and vomiting. We hypothesize that lifelong lactase activity and rapid starch digestion should be looked at as the evolutionary covalent of oral rehydration therapy.
Collapse
|
19
|
Rotavirus NSP4 Triggers Secretion of Proinflammatory Cytokines from Macrophages via Toll-Like Receptor 2. J Virol 2013; 87:11160-7. [PMID: 23926349 DOI: 10.1128/jvi.03099-12] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Nonstructural protein 4 (NSP4), encoded by rotavirus, exhibits various properties linked to viral pathogenesis, including enterotoxic activity. A recent study (O. V. Kavanagh et al., Vaccine 28:3106-3111, 2010) indicated that NSP4 also has adjuvant properties, suggesting a possible role in the innate immune response to rotavirus infection. We report here that NSP4 purified from the medium of rotavirus-infected Caco-2 cells triggers the secretion of proinflammatory cytokines from macrophage-like THP-1 cells and nitric oxide from murine RAW 264.7 cells. Secretion is accompanied by the stimulation of p38 and JNK mitogen-activated protein kinases (MAPKs) and nuclear factor NF-κB. NSP4 triggered the secretion of cytokines from murine macrophages derived from wild-type but not MyD88(-/-) or Toll-like receptor 2 (TLR2(-/-)) mice and induced secretion of interleukin-8 (IL-8) from human embryonic kidney cells transfected with TLR2 but not TLR4. Our studies identify NSP4 as a pathogen-associated molecular pattern (PAMP) encoded by rotavirus and provide a mechanism for the production of proinflammatory cytokines associated with the clinical symptoms of infection in humans and animals.
Collapse
|
20
|
Sastri NP, Pamidimukkala K, Marathahalli JR, Kaza S, Rao CD. Conformational Differences Unfold a Wide Range of Enterotoxigenic Abilities Exhibited by rNSP4 Peptides from Different Rotavirus Strains. Open Virol J 2011; 5:124-35. [PMID: 22253650 PMCID: PMC3256577 DOI: 10.2174/1874357901105010124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/18/2011] [Accepted: 09/06/2011] [Indexed: 12/28/2022] Open
Abstract
NSP4 has been recognized as the rotavirus-encoded enterotoxin. However, a few studies failed to support its diarrheagenic activity. As recombinant NSP4 (rNSP4) peptides of different lengths were used in the limited number of studies, a comparison of relative diarrheagenic potential of NSP4 from different strains could not be possible. To better understand the diarrheagenic potential of NSP4 from different strains, in this report we have evaluated the enterotoxigenic activity of the deletion mutant ΔN72 that lacks the N-terminal 72 residues and the biologically relevant ΔN112 peptide which when derived from SA11 rotavirus strain were previously shown to be highly diarrheagenic in newborn mice. Detailed comparative analysis of biochemical and biophysical properties and diarrheagenic activity of the recombinant ΔN72 peptides from seventeen different strains under identical conditions revealed wide differences among themselves in their resistance to trypsin cleavage, thioflavin T (ThT) binding, multimerization and conformation without any correlation with their diarrhea inducing abilities. These results support our previously proposed concept for the requirement of a unique conformation for optimal biological functions conferred by cooperation between the N- and C-terminal regions of the cytoplasmic tail.
Collapse
|
21
|
Gibbons TF, Storey SM, Williams CV, McIntosh A, Mitchel DM, Parr RD, Schroeder ME, Schroeder F, Ball JM. Rotavirus NSP4: Cell type-dependent transport kinetics to the exofacial plasma membrane and release from intact infected cells. Virol J 2011; 8:278. [PMID: 21645398 PMCID: PMC3129587 DOI: 10.1186/1743-422x-8-278] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 06/06/2011] [Indexed: 11/10/2022] Open
Abstract
Background Rotavirus NSP4 localizes to multiple intracellular sites and is multifunctional, contributing to RV morphogenesis, replication and pathogenesis. One function of NSP4 is the induction of early secretory diarrhea by binding surface receptors to initiate signaling events. The aims of this study were to determine the transport kinetics of NSP4 to the exofacial plasma membrane (PM), the subsequent release from intact infected cells, and rebinding to naïve and/or neighboring cells in two cell types. Methods Transport kinetics was evaluated using surface-specific biotinylation/streptavidin pull-downs and exofacial exposure of NSP4 was confirmed by antibody binding to intact cells, and fluorescent resonant energy transfer. Transfected cells similarly were monitored to discern NSP4 movement in the absence of infection or other viral proteins. Endoglycosidase H digestions, preparation of CY3- or CY5- labeled F(ab)2 fragments, confocal imaging, and determination of preferential polarized transport employed standard laboratory techniques. Mock-infected, mock-biotinylated and non-specific antibodies served as controls. Results Only full-length (FL), endoglycosidase-sensitive NSP4 was detected on the exofacial surface of two cell types, whereas the corresponding cell lysates showed multiple glycosylated forms. The C-terminus of FL NSP4 was detected on exofacial-membrane surfaces at different times in different cell types prior to its release into culture media. Transport to the PM was rapid and distinct yet FL NSP4 was secreted from both cell types at a time similar to the release of virus. NSP4-containing, clarified media from both cells bound surface molecules of naïve cells, and imaging showed secreted NSP4 from one or more infected cells bound neighboring cell membranes in culture. Preferential sorting to apical or basolateral membranes also was distinct in different polarized cells. Conclusions The intracellular transport of NSP4 to the PM, translocation across the PM, exposure of the C-terminus on the cell surface and subsequent secretion occurs via an unusual, complex and likely cell-dependent process. The exofacial exposure of the C-terminus poses several questions and suggests an atypical mechanism by which NSP4 traverses the PM and interacts with membrane lipids. Mechanistic details of the unconventional trafficking of NSP4, interactions with host-cell specific molecules and subsequent release require additional study.
Collapse
Affiliation(s)
- Thomas F Gibbons
- Department of Pathobiology Texas A&M University, TVMC, College Station, TX 77843-4467, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ousingsawat J, Mirza M, Tian Y, Roussa E, Schreiber R, Cook DI, Kunzelmann K. Rotavirus toxin NSP4 induces diarrhea by activation of TMEM16A and inhibition of Na+ absorption. Pflugers Arch 2011; 461:579-89. [PMID: 21399895 DOI: 10.1007/s00424-011-0947-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/18/2011] [Accepted: 02/21/2011] [Indexed: 01/05/2023]
Abstract
Rotavirus infection is the most frequent cause for severe diarrhea in infants, killing more than 600,000 every year. The nonstructural protein NSP4 acts as a rotavirus enterotoxin, inducing secretory diarrhea without any structural organ damage. Electrolyte transport was assessed in the colonic epithelium from pups and adult mice using Ussing chamber recordings. Western blots and immunocytochemistry was performed in intestinal tissues from wild-type and TMEM16A knockout mice. Ion channel currents were recorded using patch clamp techniques. We show that the synthetic NSP4(114-135) peptide uses multiple pro-secretory pathways to induce diarrhea, by activating the recently identified Ca2+ -activated Cl- channel TMEM16A, and by inhibiting Na+ absorption by the epithelial Na+ channel ENaC and the Na+ /glucose cotransporter SGLT1. Activation of secretion and inhibition of Na+ absorption by NSP4(114-135), respectively, could be potently suppressed by wheat germ agglutinin which probably competes with NSP4(114-135) for binding to an unknown glycolipid receptor. The present paper gives a clue as to mechanisms of rotavirus-induced diarrhea and suggests wheat germ agglutinin as a simple and effective therapy.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Toxin mediated diarrhea in the 21 century: the pathophysiology of intestinal ion transport in the course of ETEC, V. cholerae and rotavirus infection. Toxins (Basel) 2010; 2:2132-57. [PMID: 22069677 PMCID: PMC3153279 DOI: 10.3390/toxins2082132] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 08/09/2010] [Indexed: 12/31/2022] Open
Abstract
An estimated 4 billion episodes of diarrhea occur each year. As a result, 2–3 million children and 0.5–1 million adults succumb to the consequences of this major healthcare concern. The majority of these deaths can be attributed to toxin mediated diarrhea by infectious agents, such as E. coli, V. cholerae or Rotavirus. Our understanding of the pathophysiological processes underlying these infectious diseases has notably improved over the last years. This review will focus on the cellular mechanism of action of the most common enterotoxins and the latest specific therapeutic approaches that have been developed to contain their lethal effects.
Collapse
|
24
|
|
25
|
Abstract
Infectious diarrhea in calves is most commonly associated with enterotoxigenic Escherichia coli, Cryptosporidium parvum, rotavirus, coronavirus, or some combination of these pathogens. Each of these agents leads to diarrhea through either secretion or malabsorption/maldigestion, though the specific mechanisms and pathways may differ. Specific pharmacologic control and treatment are dependent on gaining a greater understanding of the pathophysiology of these organisms.
Collapse
Affiliation(s)
- D M Foster
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, 4700 Hillsborough Street, Raleigh, NC 27606, USA.
| | | |
Collapse
|
26
|
Abstract
The rotavirus is the major cause of infantile gastroenteritis. The virus infects the mature enterocytes of the villus tip of the small intestine and induces a watery diarrhea. Diarrhea can occur in the absence of histological changes in the intestine, and, conversely, the histological changes can be asymptomatic. Rotavirus decreases the activities of digestive enzymes at the apical brush border membrane and inhibits Na+ -solute cotransport systems. Accumulation of carbohydrates in the intestinal lumen as well as malabsorption of nutrients and a concomitant inhibition of water absorption can lead to a malabsorptive component of diarrhea. Since the discovery of the NSP4 enterotoxin, several hypotheses have been proposed in favour of an additional secretion component in the pathogenesis of diarrhea. Rotavirus induces a moderate net chloride secretion at the onset of the diarrhea. The mechanisms appear to different from those used by bacterial enterotoxin that cause pure secretory diarrhea. Rotavirus stimulated C1- reabsorption in villi, and failed to stimulate C1- secretion in crypt. Intestinal villi could secrete chloride as a result of rotavirus infection. The chloride secretory response is regulated by a dependant calcium signalling pathway induced by NSP4. The overall response is weak, suggesting that NSP4 may exert both secretory and subsequent antisecretory actions, hence limiting C1- secretion.
Collapse
Affiliation(s)
- M Lorrot
- Service de Pédiatrie Générale, Hôpital Robert Debré 82, Boulevard Sérurier 75019 Paris, France.
| | | |
Collapse
|
27
|
Affiliation(s)
- Adam J Moeser
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA
| | | |
Collapse
|
28
|
Beau I, Cotte-Laffitte J, Géniteau-Legendre M, Estes MK, Servin AL. An NSP4-dependant mechanism by which rotavirus impairs lactase enzymatic activity in brush border of human enterocyte-like Caco-2 cells. Cell Microbiol 2007; 9:2254-66. [PMID: 17506819 DOI: 10.1111/j.1462-5822.2007.00956.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lactase-phlorizin hydrolase (LPH, EC 3.2.1.23-62) is a brush border membrane (BBM)-associated enzyme in intestinal cells that hydrolyse lactose, the most important sugar in milk. Impairing in lactase activity during rotavirus infection has been described in diseased infants but the mechanism by which the functional lesion occurs remains unknown. We undertook a study to elucidate whether rotavirus impairs the lactase enzymatic activity in BBM of human enterocyte cells. In this study we use cultured human intestinal fully differentiated enterocyte-like Caco-2 cells to demonstrate how the lactase enzymatic activity at BBM is significantly decreased in rhesus monkey rotavirus (RRV)-infected cells. We found that the decrease in enzyme activity is not dependent of the Ca(2+)- and cAMP-dependent signalling events triggered by the virus. The LPH biosynthesis, stability, and expression of the protein at the BBM of infected cells were not modified. We provide evidence that in RRV-infected cells the kinetic of lactase enzymatic activity present at the BBM was modified. Both BBM(control) and BBM(RRV) have identical K(m) values, but hydrolyse the substrate at different rates. Thus, the BBM(RRV) exhibits almost a 1.5-fold decreased V(max) than that of BBM(control) and is therefore enzymatically less active than the latter. Our study demonstrate conclusively that the impairment of lactase enzymatic activity at the BBM of the enterocyte-like Caco-2 cells observed during rotavirus infection results from an inhibitory action of the secreted non-structural rotavirus protein NSP4.
Collapse
Affiliation(s)
- Isabelle Beau
- INSERM, UMR 756, Signalisation et Physiopathologie des Cellules Epithéliales, Châtenay-Malabry, France
| | | | | | | | | |
Collapse
|
29
|
Borghan MA, Mori Y, El-Mahmoudy AB, Ito N, Sugiyama M, Takewaki T, Minamoto N. Induction of nitric oxide synthase by rotavirus enterotoxin NSP4: implication for rotavirus pathogenicity. J Gen Virol 2007; 88:2064-2072. [PMID: 17554041 DOI: 10.1099/vir.0.82618-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Rotavirus non-structural protein (NSP) 4 can induce aqueous secretion in the gastrointestinal tract of neonatal mice through activation of an age- and Ca(2+)-dependent plasma membrane anion permeability. Accumulating evidence suggests that nitric oxide (NO) plays a role in the modulation of aqueous secretion and the barrier function of intestinal cells. This study investigated transcriptional changes in inducible NO synthase (iNOS), an enzyme responsible for NO production, after rotavirus infection in mice and after treatment of intestinal cells with NSP4. Diarrhoea was observed in 5-day-old CD-1 mice from days 1 to 3 after inoculation with 10(7) focus-forming units of different rotavirus strains. Ileal iNOS mRNA expression was induced as early as 6 h post-inoculation, before the onset of clinical diarrhoea in infected mice, and was upregulated during the course of rotavirus-induced diarrhoea. Ex vivo treatment of ilea excised from CD-1 suckling mice with NSP4 resulted in upregulation of ileal iNOS mRNA expression within 4 h. Furthermore, NSP4 was able to induce iNOS expression and NO production in murine peritoneal macrophages and RAW264.7 cells. The specificity of NSP4 inducibility was confirmed by the inhibitory effect of anti-NSP4 serum. Using a series of truncated NSP4s, the domain responsible for iNOS induction in macrophages was mapped to the reported enterotoxin domain, aa 109-135. Thus, rotavirus infection induces ileal iNOS expression in vivo and rotavirus NSP4 also induces iNOS expression in the ileum and macrophages. Together, these findings suggest that NO plays a role in rotavirus-induced diarrhoea.
Collapse
Affiliation(s)
- Mohamed A Borghan
- Laboratory of Zoonotic Diseases, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yoshio Mori
- Research Institute for Microbial Diseases, Osaka University, Japan
| | - Abu-Baker El-Mahmoudy
- Laboratory of Physiology, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Naoto Ito
- Laboratory of Zoonotic Diseases, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Makoto Sugiyama
- Laboratory of Zoonotic Diseases, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Tadashi Takewaki
- Laboratory of Physiology, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Nobuyuki Minamoto
- Laboratory of Zoonotic Diseases, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| |
Collapse
|
30
|
Lorrot M, Vasseur M. How do the rotavirus NSP4 and bacterial enterotoxins lead differently to diarrhea? Virol J 2007; 4:31. [PMID: 17376232 PMCID: PMC1839081 DOI: 10.1186/1743-422x-4-31] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Accepted: 03/21/2007] [Indexed: 01/02/2023] Open
Abstract
Rotavirus is the major cause of infantile gastroenteritis and each year causes 611,000 deaths worldwide. The virus infects the mature enterocytes of the villus tip of the small intestine and induces a watery diarrhea. Diarrhea can occur with no visible tissue damage and, conversely, the histological lesions can be asymptomatic. Rotavirus impairs activities of intestinal disaccharidases and Na+-solute symports coupled with water transport. Maldigestion of carbohydrates and their accumulation in the intestinal lumen as well as malabsorption of nutrients and a concomitant inhibition of water reabsorption can lead to a malabsorption component of diarrhea. Since the discovery of the NSP4 enterotoxin, diverse hypotheses have been proposed in favor of an additional secretion component in the pathogenesis of diarrhea. Rotavirus induces a moderate net chloride secretion at the onset of diarrhea, but the mechanisms appear to be quite different from those used by bacterial enterotoxins that cause pure secretory diarrhea. Rotavirus failed to stimulate Cl- secretion in crypt, whereas it stimulated Cl- reabsorption in villi, questioning, therefore, the origin of net Cl- secretion. A solution to this riddle was that intestinal villi do in fact secrete chloride as a result of rotavirus infection. Also, the overall chloride secretory response is regulated by a phospholipase C-dependent calcium signaling pathway induced by NSP4. However, the overall response is weak, suggesting that NSP4 may exert both secretory and subsequent anti-secretory actions, as did carbachol, hence limiting Cl- secretion. All these characteristics provide the means to make the necessary functional distinction between viral NSP4 and bacterial enterotoxins.
Collapse
Affiliation(s)
- Mathie Lorrot
- Hôpital Robert Debré, Service de Pédiatrie Générale, Paris, F-75019, France
| | - Monique Vasseur
- INSERM, UMR-S756, Université Paris-Sud 11, Faculté de Pharmacie, Châtenay-Malabry, F-92296, France
| |
Collapse
|
31
|
Beau I, Berger A, Servin AL. Rotavirus impairs the biosynthesis of brush-border-associated dipeptidyl peptidase IV in human enterocyte-like Caco-2/TC7 cells. Cell Microbiol 2007; 9:779-89. [PMID: 17081193 DOI: 10.1111/j.1462-5822.2006.00827.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Rotavirus is the leading cause of severe dehydrating diarrhoea in infants and young children worldwide. This virus infects mature enterocytes in the small intestine, and induces structural and functional damage. In the present study, we have identified a new mechanism by which rotavirus impairs a brush border-associated intestinal protein. We show that infection of enterocyte-like Caco-2/TC7 cells by rhesus monkey rotavirus (RRV) impairs the biosynthesis of dipeptidyl peptidase IV (DPP IV), an important hydrolase in the digestion of dietary proline-rich proteins. We show that the enzyme activity of DPP IV was reduced, and that rearrangements of the protein occurred at the apical domain of the RRV-infected cells. Using pulse-chase experiments and cell surface immunoprecipitation, we have demonstrated that RRV infection did not affect the stability or apical targeting of DPP IV, but did induce a dramatic decrease in its biosynthesis. Using quantitative RT-PCR, we showed that RRV had no effect on the level of expression of DPP IV mRNA, suggesting that the observed decrease in the biosynthesis of the protein is related to an effect of the virus at the translational level.
Collapse
Affiliation(s)
- Isabelle Beau
- Institut National de la Santé et de la Recherche Médicale, Université Paris XI, UMR-S 756, Signalisation et Physiopathologie des Cellules Epithéliales, Faculté de Pharmacie, Châtenay-Malabry, F-92296 France
| | | | | |
Collapse
|
32
|
Iovane G, Pisanelli G, Pagnini U. Rotavirus diarrhoea in Buffaloes: epidemiology, pathogenesys and prophilaxis. ITALIAN JOURNAL OF ANIMAL SCIENCE 2007. [DOI: 10.4081/ijas.2007.s2.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Lorrot M, Vasseur M. Rotavirus NSP4 114-135 peptide has no direct, specific effect on chloride transport in rabbit brush-border membrane. Virol J 2006; 3:94. [PMID: 17101044 PMCID: PMC1657008 DOI: 10.1186/1743-422x-3-94] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Accepted: 11/13/2006] [Indexed: 12/24/2022] Open
Abstract
The direct effect of the rotavirus NSP4114-135 and Norovirus NV464-483 peptides on 36Cl uptake was studied by using villus cell brush border membrane (BBM) isolated from young rabbits. Both peptides inhibited the Cl-/H+ symport activity about equally and partially. The interaction involved one peptide-binding site per carrier unit. Whereas in vitro NSP4114-135 caused nonspecific inhibition of the Cl-/H+ symporter, the situation in vivo is different. Because rotavirus infection in young rabbits accelerated both Cl- influx and Cl- efflux rates across villi BBM without stimulating Cl- transport in crypt BBM, we conclude that the NSP4114-135 peptide, which causes diarrhea in young rodents, did not have any direct, specific effect on either intestinal absorption or secretion of chloride. The lack of direct effect of NSP4 on chloride transport strengthens the hypothesis that NSP4 would trigger signal transduction pathways to enhance net chloride secretion at the onset of rotavirus diarrhea.
Collapse
Affiliation(s)
- Mathie Lorrot
- Hôpital Robert Debré, Service de Pédiatrie Générale, Paris, F-75019, France
| | - Monique Vasseur
- INSERM, UMR 756, Université de Paris XI, Faculté de Pharmacie, Châtenay-Malabry, F-92296, France
| |
Collapse
|
34
|
Parr RD, Storey SM, Mitchell DM, McIntosh AL, Zhou M, Mir KD, Ball JM. The rotavirus enterotoxin NSP4 directly interacts with the caveolar structural protein caveolin-1. J Virol 2006; 80:2842-54. [PMID: 16501093 PMCID: PMC1395425 DOI: 10.1128/jvi.80.6.2842-2854.2006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Accepted: 12/13/2005] [Indexed: 11/20/2022] Open
Abstract
Rotavirus nonstructural protein 4 (NSP4) is known to function as an intracellular receptor at the endoplasmic reticulum (ER) critical to viral morphogenesis and is the first characterized viral enterotoxin. Exogenously added NSP4 induces diarrhea in rodent pups and stimulates secretory chloride currents across intestinal segments as measured in Ussing chambers. Circular dichroism studies further reveal that intact NSP4 and the enterotoxic peptide (NSP4(114-135)) that is located within the extended, C-terminal amphipathic helix preferentially interact with caveola-like model membranes. We now show colocalization of NSP4 and caveolin-1 in NSP4-transfected and rotavirus-infected mammalian cells in reticular structures surrounding the nucleus (likely ER), in the cytosol, and at the cell periphery by laser scanning confocal microscopy. A direct interaction between NSP4 residues 112 to 140 and caveolin-1 was determined by the Pro-Quest yeast two-hybrid system with full-length NSP4 and seven overlapping deletion mutants as bait, caveolin-1 as prey, and vice versa. Coimmunoprecipitation of NSP4-caveolin-1 complexes from rotavirus-infected mammalian cells demonstrated that the interaction occurs during viral infection. Finally, binding of caveolin-1 from mammalian cell lysates to Sepharose-bound, NSP4-specific synthetic peptides confirmed the yeast two-hybrid data and further delineated the binding domain to amino acids 114 to 135. We propose that the association of NSP4 and caveolin-1 contributes to NSP4 intracellular trafficking from the ER to the cell surface and speculate that exogenously added NSP4 stimulates signaling molecules located in caveola microdomains.
Collapse
Affiliation(s)
- Rebecca D Parr
- Department of Pathobiology, Texas A&M University 4467, College Station, Texas 77843, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Canani RB, De Marco G, Passariello A, Buccigrossi V, Ruotolo S, Bracale I, Porcaro F, Bifulco G, Guarino A. Inhibitory effect of HIV-1 Tat protein on the sodium-D-glucose symporter of human intestinal epithelial cells. AIDS 2006; 20:5-10. [PMID: 16327313 DOI: 10.1097/01.aids.0000198088.85572.68] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The pathophysiology of HIV-1-related intestinal dysfunction is largely unknown. We previously found that the transactivator factor peptide (Tat) produced by HIV-1 induces ion secretion and inhibits cell proliferation in human enterocytes. Because sugar malabsorption is a frequent feature in AIDS patients, we evaluated whether Tat inhibits intestinal glucose absorption. DESIGN AND METHODS We measured Na-D-glucose symporter (SGLT-1) activity and determined its phenotypic expression in Caco-2 cells, in the presence and absence of Tat, in uptake experiments using a non-metabolized radiolabelled glucose analogue, and by western blot analysis, respectively. alpha-Tubulin staining was used to study the effects exerted by Tat on cell structure. RESULTS Tat dose dependently inhibited glucose uptake by human enterocytes. This effect was prevented by anti-Tat polyclonal antibodies and by L-type Ca channels agonist Bay K8644. Western blot analysis of cellular lysates and brush-border membrane preparations showed that Tat induced SGLT-1 missorting. Tat also caused a dramatic decrease in alpha-tubulin staining, which indicates dysruption of the cytoskeleton organization. CONCLUSIONS Tat acutely impairs intestinal glucose absorption through SGLT-1 missorting. This result indicates that Tat is directly involved in AIDS-associated intestinal dysfunction.
Collapse
|
36
|
Molecular Virology of Enteric Viruses (with Emphasis on Caliciviruses). VIRUSES IN FOODS 2006:43-100. [PMCID: PMC7120911 DOI: 10.1007/0-387-29251-9_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
|
37
|
Bányai K, Forgách P, Erdélyi K, Martella V, Bogdán A, Hocsák E, Havasi V, Melegh B, Szucs G. Identification of the novel lapine rotavirus genotype P[22] from an outbreak of enteritis in a Hungarian rabbitry. Virus Res 2005; 113:73-80. [PMID: 15936106 DOI: 10.1016/j.virusres.2005.03.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 03/29/2005] [Accepted: 03/29/2005] [Indexed: 11/26/2022]
Abstract
Application of improved molecular techniques in the detection and characterization of rotavirus strains has led to the recent description of several new combinations, specificities, and genetic variants of the outer capsid genes, VP7 and VP4. In spite of the enormous diversity of mammalian rotavirus strains, the few lapine rotaviruses characterized to date, appear to carry a narrow range of such antigen combinations; only P[14], G3 and, based on a more recent study, P[22], G3 rotaviruses have proved to be epidemiologically important in rabbits. In the present study, we characterized a lapine group A rotavirus with a super-short electropherotype detected in an outbreak of fatal enteritis in a Hungarian commercial rabbitry. Based on sequence and phylogenetic analysis of the VP7, VP4, and NSP4 genes, our lapine strain is a P[22], G3 rotavirus that carries the NSP4 genotype shared by most lapine rotaviruses. Although the P[22] VP4 specificity has been newly identified, the relatively high sequence variation between our strain and those identified in Italy (89.1-90.4% nucleotide identity; region VP8*) implies that these strains diversified far before they were described for the first time, strongly suggesting that this genotype may have circulated in rabbitries or in nature without prior detection. We conclude that genotype P[22] lapine rotaviruses show a wider geographical dispersal than previously thought, although understanding their true epidemiological significance needs further investigation.
Collapse
Affiliation(s)
- Krisztián Bányai
- Regional Laboratory of Virology, Baranya County Institute of State Public Health Service, Szabadság út 7, H-7623 Pécs, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ball JM, Mitchell DM, Gibbons TF, Parr RD. Rotavirus NSP4: a multifunctional viral enterotoxin. Viral Immunol 2005; 18:27-40. [PMID: 15802952 DOI: 10.1089/vim.2005.18.27] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Judith M Ball
- Department of Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, 4467 TAMU, College Station, TX 77843, USA.
| | | | | | | |
Collapse
|
39
|
Carter MJ. Enterically infecting viruses: pathogenicity, transmission and significance for food and waterborne infection. J Appl Microbiol 2005; 98:1354-80. [PMID: 15916649 DOI: 10.1111/j.1365-2672.2005.02635.x] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- M J Carter
- School of Biomedical and Molecular Sciences, University of Surrey, Guildford GU27XH, UK.
| |
Collapse
|
40
|
Rodríguez-Díaz J, Montava R, García-Díaz A, Buesa J. Humoral immune response to rotavirus NSP4 enterotoxin in Spanish children. J Med Virol 2005; 77:317-22. [PMID: 16121377 DOI: 10.1002/jmv.20450] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The rotavirus non-structural protein 4 (NSP4) has been shown to play a crucial role in rotavirus-induced diarrhea, acting as a viral enterotoxin. It has also been demonstrated that antibody to NSP4 can reduce the severity of rotavirus-induced diarrhea in newborn mice. Two recombinant baculoviruses, expressing the NSP4 protein from the SA11 and Wa rotavirus strains, genotypes A and B, respectively, were used to produce and purify these glycoproteins, which were applied as antigen in an enzyme-linked immunosorbent assay (ELISA) to test the specific antibody response to NSP4 in human sera. Serum samples from 30 children convalescing from a rotavirus infection, from 54 healthy children under 5-years-old, and from 49 adults were tested to determine the presence of antibodies to the viral enterotoxin and to rotavirus structural proteins. Seventy percent of the sera from rotavirus-infected children showed an IgG antibody response to either one or both NSP4 proteins used in this study, although the response was weak. However, IgG antibodies towards either one or both NSP4 proteins were only detected in 26% of the non-convalescent healthy children and in only 18% of the adults. No serum IgA antibodies towards NSP4 were found in this study. IgG antibody recognition of the NSP4 protein from the SA11 and Wa rotavirus strains was not always heterotypic.
Collapse
Affiliation(s)
- Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, Hospital Clínico Universitario, University of Valencia, Valencia, Spain
| | | | | | | |
Collapse
|
41
|
Huang H, Schroeder F, Estes MK, McPherson T, Ball JM. Interaction(s) of rotavirus non-structural protein 4 (NSP4) C-terminal peptides with model membranes. Biochem J 2004; 380:723-33. [PMID: 15012630 PMCID: PMC1224213 DOI: 10.1042/bj20031789] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Revised: 03/09/2004] [Accepted: 03/10/2004] [Indexed: 01/18/2023]
Abstract
Rotavirus is the major cause of dehydrating gastroenteritis in children and young animals. NSP4 (non-structural protein 4), a rotaviral non-structural glycoprotein and a peptide NSP4(114-135) (DKLTTREIEQVELLKRIYDKLT), corresponding to NSP4 amino acids 114-135, induce diarrhoeal disease in a neonatal mouse model and interact with model membranes that mimic caveolae. Correlation of the mechanisms of diarrhoea induction and membrane interactions by NSP4 protein and peptide remain unclear. Several additional NSP4 peptides were synthesized and their interactions with membranes studied by (i) CD, (ii) a filtration-binding assay and (iii) a fluorescent molecule leakage assay. Model membranes that varied in lipid compositions and radius of curvature were utilized to determine the compositional and structural requirements for optimal interaction with the peptides of NSP4. Similar to the intact protein and NSP4(114-135), peptides overlapping residues 114-135 had significantly higher affinities to membranes rich in negatively charged lipids, rich in cholesterol and with a high radius of curvature. In the leakage assay, small and large unilamellar vesicles loaded with the fluorophore/quencher pair 8-aminonaphthalene-1,3,6-trisulphonic acid disodium salt/p -xylene-bis-pyridinium bromide were incubated with the NSP4 peptides and monitored for membrane disruption by lipid reorganization or by pore formation. At a peptide concentration of 15 microM, none of the NSP4 peptides caused leakage. These results confirm that NSP4 interacts with caveolae-like membranes and the alpha-helical region of NSP4(114-135) comprises a membrane interaction domain that does not induce membrane disruption at physiological concentrations.
Collapse
Affiliation(s)
- Huan Huang
- Department of Pharmacology and Physiology, Texas A&M University, TVMC, College Station, TX 77843-4466, USA
| | | | | | | | | |
Collapse
|
42
|
Kunzelmann K, McMorran B. First Encounter: How Pathogens Compromise Epithelial Transport. Physiology (Bethesda) 2004; 19:240-4. [PMID: 15381751 DOI: 10.1152/physiol.00015.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pathogenic organisms trigger numerous signaling pathways that ultimately lead to drastic changes in physiological functions. Apart from altering structure and function of the epithelial tight junction barrier and activating inflammatory cascades, they induce changes in fluid and electrolyte transport. Pathogens do so by activating or by inhibiting ion channels and transporters, and the result might be to their benefit or to their disadvantage.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, D-93053 Regensburg, Germany.
| | | |
Collapse
|
43
|
Affiliation(s)
- Robert F Ramig
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
44
|
Boshuizen JA, Rossen JWA, Sitaram CK, Kimenai FFP, Simons-Oosterhuis Y, Laffeber C, Büller HA, Einerhand AWC. Rotavirus enterotoxin NSP4 binds to the extracellular matrix proteins laminin-beta3 and fibronectin. J Virol 2004; 78:10045-53. [PMID: 15331737 PMCID: PMC514988 DOI: 10.1128/jvi.78.18.10045-10053.2004] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rotavirus is the most important cause of viral gastroenteritis and dehydrating diarrhea in young children. Rotavirus nonstructural protein 4 (NSP4) is an enterotoxin that was identified as an important agent in symptomatic rotavirus infection. To identify cellular proteins that interact with NSP4, a two-hybrid technique with Saccharomyces cerevisiae was used. NSP4 cDNA, derived from the human rotavirus strain Wa, was cloned into the yeast shuttle vector pGBKT7. An intestinal cDNA library derived from Caco-2 cells cloned into the yeast shuttle vector pGAD10 was screened for proteins that interact with NSP4. Protein interactions were confirmed in vivo by coimmunoprecipitation and immunohistochemical colocalization. After two-hybrid library screening, we repeatedly isolated cDNAs encoding the extracellular matrix (ECM) protein laminin-beta3 (amino acids [aa] 274 to 878) and a cDNA encoding the ECM protein fibronectin (aa 1755 to 1884). Using deletion mutants of NSP4, we mapped the region of interaction with the ECM proteins between aa 87 and 145. Deletion analysis of laminin-beta3 indicated that the region comprising aa 726 to 875 of laminin-beta3 interacts with NSP4. Interaction of NSP4 with either laminin-beta3 or fibronectin was confirmed by coimmunoprecipitation. NSP4 was present in infected enterocytes and in the basement membrane (BM) of infected neonatal mice and colocalized with laminin-beta3, indicating a physiological interaction. In conclusion, two-hybrid screening with NSP4 yielded two potential target proteins, laminin-beta3 and fibronectin, interacting with the enterotoxin NSP4. The release of NSP4 from the basal side of infected epithelial cells and the subsequent binding to ECM proteins localized at the BM may signify a new mechanism by which rotavirus disease is established.
Collapse
Affiliation(s)
- J A Boshuizen
- Laboratory of Pediatrics, Pediatric Gastroenterology & Nutrition, Erasmus MC, Rm. Ee1571A, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Boshuizen JA, Reimerink JHJ, Korteland-van Male AM, van Ham VJJ, Koopmans MPG, Büller HA, Dekker J, Einerhand AWC. Changes in small intestinal homeostasis, morphology, and gene expression during rotavirus infection of infant mice. J Virol 2004; 77:13005-16. [PMID: 14645557 PMCID: PMC296055 DOI: 10.1128/jvi.77.24.13005-13016.2003] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Rotavirus is the most important cause of infantile gastroenteritis. Since in vivo mucosal responses to a rotavirus infection thus far have not been extensively studied, we related viral replication in the murine small intestine to alterations in mucosal structure, epithelial cell homeostasis, cellular kinetics, and differentiation. Seven-day-old suckling BALB/c mice were inoculated with 2 x 10(4) focus-forming units of murine rotavirus and were compared to mock-infected controls. Diarrheal illness and viral shedding were recorded, and small intestinal tissue was evaluated for rotavirus (NSP4 and structural proteins)- and enterocyte-specific (lactase, SGLT1, and L-FABP) mRNA and protein expression. Morphology, apoptosis, proliferation, and migration were evaluated (immuno)histochemically. Diarrhea was observed from days 1 to 5 postinfection, and viral shedding was observed from days 1 to 10. Two peaks of rotavirus replication were observed at 1 and 4 days postinfection. Histological changes were characterized by the accumulation of vacuolated enterocytes. Strikingly, the number of vacuolated cells exceeded the number of cells in which viral replication was detectable. Apoptosis and proliferation were increased from days 1 to 7, resulting in villous atrophy. Epithelial cell turnover was significantly higher (<4 days) than that observed in controls (7 days). Since epithelial renewal occurred within 4 days, the second peak of viral replication was most likely caused by infection of newly synthesized cells. Expression of enterocyte-specific genes was downregulated in infected cells at mRNA and protein levels starting as early as 6 h after infection. In conclusion, we show for the first time that rotavirus infection induces apoptosis in vivo, an increase in epithelial cell turnover, and a shutoff of gene expression in enterocytes showing viral replication. The shutoff of enterocyte-specific gene expression, together with the loss of mature enterocytes through apoptosis and the replacement of these cells by less differentiated dividing cells, likely leads to a defective absorptive function of the intestinal epithelium, which contributes to rotavirus pathogenesis.
Collapse
Affiliation(s)
- Jos A Boshuizen
- Laboratoryof Pediatrics, Pediatric Gastroenterology, and Nutrition, Erasmus MC/ Sophia, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
46
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2003; 11:1670-1673. [DOI: 10.11569/wcjd.v11.i11.1670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
47
|
II, 8. Effects of rotavirus infection on the structure and functions of intestinal cells. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s0168-7069(03)09015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
48
|
II, 6.The rotavirus NSP4 enterotoxin: Current status and challenges. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s0168-7069(03)09013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
49
|
Mori Y, Borgan MA, Ito N, Sugiyama M, Minamoto N. Sequential analysis of nonstructural protein NSP4s derived from Group A avian rotaviruses. Virus Res 2002; 89:145-51. [PMID: 12367757 DOI: 10.1016/s0168-1702(02)00112-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We determined the NSP4 sequences of turkey rotavirus strains Ty-1 and Ty-3 and a chicken rotavirus, strain Ch-1, and compared these sequences with those of a pigeon rotavirus, strain PO-13, and mammalian rotaviruses. The turkey strains and PO-13 were found to be closely related (90-97% homologies). Ch-1 NSP4 was distinctly different from other avian rotavirus NSP4s, with 78-79% homologies. The NSP4 sequences of avian rotaviruses were found to be 6-7 amino acids shorter than those of all mammalian strains and to have considerably low identities (31-37%) with them. Therefore, it seems highly likely that the NSP4 genes of avian rotaviruses are classified into two NSP4 genotypes distinct from those of mammalian rotaviruses. The enterotoxin domain in NSP4 is conserved in terms of its sequential and structural properties despite extremely low homologies in the full lengths of NSP4s in avian and mammalian rotaviruses.
Collapse
Affiliation(s)
- Yoshio Mori
- Department of Veterinary Public Health, Faculty of Agriculture, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | | | | | | | | |
Collapse
|
50
|
Mori Y, Borgan MA, Ito N, Sugiyama M, Minamoto N. Diarrhea-inducing activity of avian rotavirus NSP4 glycoproteins, which differ greatly from mammalian rotavirus NSP4 glycoproteins in deduced amino acid sequence in suckling mice. J Virol 2002; 76:5829-34. [PMID: 11992013 PMCID: PMC137017 DOI: 10.1128/jvi.76.11.5829-5834.2002] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Avian rotavirus NSP4 glycoproteins expressed in Escherichia coli acted as enterotoxins in suckling mice, as did mammalian rotavirus NSP4 glycoproteins, despite great differences in the amino acid sequences. The enterotoxin domain of PO-13 NSP4 exists in amino acid residues 109 to 135, a region similar to that reported in SA11 NSP4.
Collapse
Affiliation(s)
- Yoshio Mori
- Department of Veterinary Public Health, Faculty of Agriculture, Gifu University, Gifu 501-1193, Japan
| | | | | | | | | |
Collapse
|