Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Gastroenterology ; 165(4): 986-998.e11, 2023 10.
Article in English | MEDLINE | ID: mdl-37429363

ABSTRACT

BACKGROUND & AIMS: Acute diarrheal diseases are the second most common cause of infant mortality in developing countries. This is contributed to by lack of effective drug therapy that shortens the duration or lessens the volume of diarrhea. The epithelial brush border sodium (Na+)/hydrogen (H+) exchanger 3 (NHE3) accounts for a major component of intestinal Na+ absorption and is inhibited in most diarrheas. Because increased intestinal Na+ absorption can rehydrate patients with diarrhea, NHE3 has been suggested as a potential druggable target for drug therapy for diarrhea. METHODS: A peptide (sodium-hydrogen exchanger 3 stimulatory peptide [N3SP]) was synthesized to mimic the part of the NHE3 C-terminus that forms a multiprotein complex that inhibits NHE3 activity. The effect of N3SP on NHE3 activity was evaluated in NHE3-transfected fibroblasts null for other plasma membrane NHEs, a human colon cancer cell line that models intestinal absorptive enterocytes (Caco-2/BBe), human enteroids, and mouse intestine in vitro and in vivo. N3SP was delivered into cells via a hydrophobic fluorescent maleimide or nanoparticles. RESULTS: N3SP uptake stimulated NHE3 activity at nmol/L concentrations under basal conditions and partially reversed the reduced NHE3 activity caused by elevated adenosine 3',5'-cyclic monophosphate, guanosine 3',5'-cyclic monophosphate, and Ca2+ in cell lines and in in vitro mouse intestine. N3SP also stimulated intestinal fluid absorption in the mouse small intestine in vivo and prevented cholera toxin-, Escherichia coli heat-stable enterotoxin-, and cluster of differentiation 3 inflammation-induced fluid secretion in a live mouse intestinal loop model. CONCLUSIONS: These findings suggest pharmacologic stimulation of NHE3 activity as an efficacious approach for the treatment of moderate/severe diarrheal diseases.


Subject(s)
Enterotoxins , Sodium-Hydrogen Exchangers , Mice , Animals , Humans , Sodium-Hydrogen Exchanger 3/metabolism , Enterotoxins/pharmacology , Enterotoxins/metabolism , Caco-2 Cells , Sodium-Hydrogen Exchangers/metabolism , Enterocytes/metabolism , Sodium/metabolism , Diarrhea/drug therapy , Diarrhea/prevention & control , Diarrhea/chemically induced , Peptides/adverse effects , Microvilli/metabolism
2.
J Autoimmun ; 134: 102961, 2023 01.
Article in English | MEDLINE | ID: mdl-36470208

ABSTRACT

Regulatory T cell (Treg) therapy is a promising strategy to treat inflammatory bowel disease (IBD). Data from animal models has shown that Tregs specific for intestinal antigens are more potent than polyclonal Tregs at inhibiting colitis. Flagellins, the major structural proteins of bacterial flagella, are immunogenic antigens frequently targeted in IBD subjects, leading to the hypothesis that flagellin-specific Tregs could be an effective cell therapy for IBD. We developed a novel chimeric antigen receptor (CAR) specific for flagellin derived from Escherichia coli H18 (FliC). We used this CAR to confer FliC-specificity to human Tregs and investigated their therapeutic potential. FliC-CAR Tregs were activated by recombinant FliC protein but not a control flagellin protein, demonstrating CAR specificity and functionality. In a humanized mouse model, expression of the FliC-CAR drove preferential migration to the colon and expression of the activation marker PD1. In the presence of recombinant FliC protein in vitro, FliC-CAR Tregs were significantly more suppressive than control Tregs and promoted the establishment of colon-derived epithelial cell monolayers. These results demonstrate the potential of FliC-CAR Tregs to treat IBD and more broadly show the therapeutic potential of CARs targeting microbial-derived antigens.


Subject(s)
Inflammatory Bowel Diseases , Receptors, Chimeric Antigen , Animals , Mice , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Flagellin/metabolism , Recombinant Proteins/metabolism , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/metabolism , T-Lymphocytes, Regulatory
3.
FASEB J ; 34(12): 15922-15945, 2020 12.
Article in English | MEDLINE | ID: mdl-33047400

ABSTRACT

Enterotoxigenic Bacteroides fragilis (ETBF) is a commensal bacterium of great importance to human health due to its ability to induce colitis and cause colon tumor formation in mice through the production of B. fragilis toxin (BFT). The formation of tumors is dependent on a pro-inflammatory signaling cascade, which begins with the disruption of epithelial barrier integrity through cleavage of E-cadherin. Here, we show that BFT increases levels of glucosylceramide, a vital intestinal sphingolipid, both in mice and in colon organoids (colonoids) generated from the distal colons of mice. When colonoids are treated with BFT in the presence of an inhibitor of glucosylceramide synthase (GCS), the enzyme responsible for generating glucosylceramide, colonoids become highly permeable, lose structural integrity, and eventually burst, releasing their contents into the extracellular matrix. By increasing glucosylceramide levels in colonoids via an inhibitor of glucocerebrosidase (GBA, the enzyme that degrades glucosylceramide), colonoid permeability was reduced, and bursting was significantly decreased. In the presence of BFT, pharmacological inhibition of GCS caused levels of tight junction protein 1 (TJP1) to decrease. However, when GBA was inhibited, TJP1 levels remained stable, suggesting that BFT-induced production of glucosylceramide helps to stabilize tight junctions. Taken together, our data demonstrate a glucosylceramide-dependent mechanism by which the colon epithelium responds to BFT.


Subject(s)
Bacterial Toxins/toxicity , Bacteroides fragilis/metabolism , Colon/drug effects , Epithelial Cells/drug effects , Glucosylceramides/metabolism , Metalloendopeptidases/toxicity , Signal Transduction/drug effects , Animals , Colitis/chemically induced , Colitis/metabolism , Colon/metabolism , Epithelial Cells/metabolism , Glucosylceramidase/metabolism , Glucosyltransferases/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Permeability/drug effects , Zonula Occludens-1 Protein/metabolism
4.
Annu Rev Physiol ; 79: 291-312, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28192061

ABSTRACT

The lack of accessibility to normal and diseased human intestine and the inability to separate the different functional compartments of the intestine even when tissue could be obtained have held back the understanding of human intestinal physiology. Clevers and his associates identified intestinal stem cells and established conditions to grow "mini-intestines" ex vivo in differentiated and undifferentiated conditions. This pioneering work has made a new model of the human intestine available and has begun making contributions to the understanding of human intestinal transport in normal physiologic conditions and the pathophysiology of intestinal diseases. However, this model is reductionist and lacks many of the complexities of normal intestine. Consequently, it is not yet possible to predict how great the advances using this model will be for understanding human physiology and pathophysiology, nor how the model will be modified to include multiple other intestinal cell types and physical forces necessary to more closely approximate normal intestine. This review describes recent studies using mini-intestines, which have readdressed previously established models of normal intestinal transport physiology and newly examined intestinal pathophysiology. The emphasis is on studies with human enteroids grown either as three-dimensional spheroids or two-dimensional monolayers. In addition, comments are provided on mouse studies in cases when human studies have not yet been described.


Subject(s)
Intestinal Diseases/pathology , Intestines/pathology , Intestines/physiology , Animals , Cell Differentiation/physiology , Humans , Models, Biological , Stem Cells/pathology , Stem Cells/physiology
5.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32631917

ABSTRACT

Symptomatic and asymptomatic infection with the diarrheal pathogen enteroaggregative Escherichia coli (EAEC) is associated with growth faltering in children in developing settings. The mechanism of this association is unknown, emphasizing a need for better understanding of the interactions between EAEC and the human gastrointestinal mucosa. In this study, we investigated the role of the aggregative adherence fimbriae II (AAF/II) in EAEC adherence and pathogenesis using human colonoids and duodenal enteroids. We found that a null mutant in aafA, the major subunit of AAF/II, adhered significantly less than wild-type (WT) EAEC strain 042, and adherence was restored in a complemented strain. Immunofluorescence confocal microscopy of differentiated colonoids, which produce an intact mucus layer comprised of the secreted mucin MUC2, revealed bacteria at the epithelial surface and within the MUC2 layer. The WT strain adhered to the epithelial surface, whereas the aafA deletion strain remained within the MUC2 layer, suggesting that the presence or absence of AAF/II determines both the abundance and location of EAEC adherence. In order to determine the consequences of EAEC adherence on epithelial barrier integrity, colonoid monolayers were exposed to EAEC constructs expressing or lacking aafA Colonoids infected with WT EAEC had significantly decreased epithelial resistance, an effect that required AAF/II, suggesting that binding of EAEC to the epithelium is necessary to impair barrier function. In summary, we show that production of AAF/II is critical for adherence and barrier disruption in human colonoids, suggesting a role for this virulence factor in EAEC colonization of the gastrointestinal mucosa.


Subject(s)
Adhesins, Escherichia coli/immunology , Epithelial Cells/microbiology , Escherichia coli/immunology , Fimbriae, Bacterial/immunology , Host Microbial Interactions/immunology , Organoids/microbiology , Adhesins, Escherichia coli/genetics , Bacterial Adhesion , Colon/immunology , Colon/metabolism , Colon/microbiology , Colony Count, Microbial , Duodenum/immunology , Duodenum/metabolism , Duodenum/microbiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Fimbriae, Bacterial/genetics , Gene Deletion , Gene Expression Regulation , Genetic Complementation Test , Host Microbial Interactions/genetics , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mucin-2/genetics , Mucin-2/immunology , Organoids/immunology , Organoids/metabolism , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 114(4): E570-E579, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069942

ABSTRACT

The intestinal epithelium can limit enteric pathogens by producing antiviral cytokines, such as IFNs. Type I IFN (IFN-α/ß) and type III IFN (IFN-λ) function at the epithelial level, and their respective efficacies depend on the specific pathogen and site of infection. However, the roles of type I and type III IFN in restricting human enteric viruses are poorly characterized as a result of the difficulties in cultivating these viruses in vitro and directly obtaining control and infected small intestinal human tissue. We infected nontransformed human intestinal enteroid cultures from multiple individuals with human rotavirus (HRV) and assessed the host epithelial response by using RNA-sequencing and functional assays. The dominant transcriptional pathway induced by HRV infection is a type III IFN-regulated response. Early after HRV infection, low levels of type III IFN protein activate IFN-stimulated genes. However, this endogenous response does not restrict HRV replication because replication-competent HRV antagonizes the type III IFN response at pre- and posttranscriptional levels. In contrast, exogenous IFN treatment restricts HRV replication, with type I IFN being more potent than type III IFN, suggesting that extraepithelial sources of type I IFN may be the critical IFN for limiting enteric virus replication in the human intestine.


Subject(s)
Interferons/genetics , Intestine, Small/immunology , Rotavirus Infections/genetics , Animals , Cell Line , Chlorocebus aethiops , Humans , Immunity, Innate , Interferons/immunology , Rotavirus/physiology , Rotavirus Infections/immunology , Sequence Analysis, RNA , Virus Replication
7.
Infect Immun ; 87(4)2019 04.
Article in English | MEDLINE | ID: mdl-30642900

ABSTRACT

The enteric pathogen Shigella is one of the leading causes of moderate-to-severe diarrhea and death in young children in developing countries. Transformed cell lines and animal models have been widely used to study Shigella pathogenesis. In addition to altered physiology, transformed cell lines are composed of a single cell type that does not sufficiently represent the complex multicellular environment of the human colon. Most available animal models do not accurately mimic human disease. The human intestinal enteroid model, derived from LGR5+ stem cell-containing intestinal crypts from healthy subjects, represents a technological leap in human gastrointestinal system modeling and provides a more physiologically relevant system that includes multiple cell types and features of the human intestine. We established the utility of this model for studying basic aspects of Shigella pathogenesis and host responses. In this study, we show that Shigellaflexneri is capable of infecting and replicating intracellularly in human enteroids derived from different segments of the intestine. Apical invasion by S. flexneri is very limited but increases ∼10-fold when enteroids are differentiated to include M cells. Invasion via the basolateral surface was at least 2-log10 units more efficient than apical infection. Increased secretion of interleukin-8 and higher expression levels of the mucin glycoprotein Muc2 were observed in the enteroids following S. flexneri infection. The human enteroid model promises to bridge some of the gaps between traditional cell culture, animal models, and human infection.


Subject(s)
Dysentery, Bacillary/microbiology , Intestines/cytology , Organoids/microbiology , Shigella flexneri/physiology , Cells, Cultured , Humans , Intestines/microbiology , Models, Biological , Organoids/growth & development , Organoids/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Shigella flexneri/genetics , Shigella flexneri/growth & development , Shigella flexneri/pathogenicity , Stem Cells/cytology , Stem Cells/metabolism , Virulence
8.
J Biol Chem ; 292(20): 8279-8290, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28283572

ABSTRACT

NHE3 directly binds Na+/H+ exchanger regulatory factor (NHERF) family scaffolding proteins that are required for many aspects of NHE3 regulation. The NHERFs bind both to an internal region (amino acids 586-660) of the NHE3 C terminus and to the NHE3 C-terminal four amino acids. The internal NHERF-binding region contains both putative Class I (-592SAV-) and Class II (-595CLDM-) PDZ-binding motifs (PBMs). Point mutagenesis showed that only the Class II motif contributes to NHERF binding. In this study, the roles in regulation of NHE3 activity of these two PBMs were investigated, revealing the following findings. 1) Interaction occurred between these binding sites because mutation of either removed nearly all NHERF binding. 2) Mutations in either significantly reduced basal NHE3 activity. Total and percent plasma membrane (PM) NHE3 protein expression was reduced in the C-terminal but not in the internal PBD mutation. 3) cGMP- and Ca2+-mediated inhibition of NHE3 was impaired in both the internal and the C-terminal PBM mutations. 4) There was a significant reduction in half-life of the PM pool of NHE3 in only the internal PBM mutation but no change in total NHE3 half-life in either. 5) There were some differences in NHE3-associating proteins in the two PBM mutations. In conclusion, NHE3 binds to NHERF proteins via both an internal Class II PBM and C-terminal Class I PBM, which interact. The former determines NHE3 stability in the PM, and the latter determines total expression and percent PM expression.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Cyclic GMP/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Motifs , Cell Line , Cell Membrane/genetics , Cyclic GMP/genetics , Humans , Mutation , PDZ Domains , Phosphoproteins/genetics , Protein Binding/physiology , Protein Stability , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics
9.
FASEB J ; 31(2): 751-760, 2017 02.
Article in English | MEDLINE | ID: mdl-27871064

ABSTRACT

Secretory diarrheas caused by bacterial enterotoxins, including cholera and traveler's diarrhea, remain a major global health problem. Inappropriate activation of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel occurs in these diarrheas. We previously reported that the benzopyrimido-pyrrolo-oxazinedione (R)-BPO-27 inhibits CFTR chloride conductance with low-nanomolar potency. Here, we demonstrate using experimental mouse models and human enterocyte cultures the potential utility of (R)-BPO-27 for treatment of secretory diarrheas caused by cholera and Escherichia coli enterotoxins. (R)-BPO-27 fully blocked CFTR chloride conductance in epithelial cell cultures and intestine after cAMP agonists, cholera toxin, or heat-stable enterotoxin of E. coli (STa toxin), with IC50 down to ∼5 nM. (R)-BPO-27 prevented cholera toxin and STa toxin-induced fluid accumulation in small intestinal loops, with IC50 down to 0.1 mg/kg. (R)-BPO-27 did not impair intestinal fluid absorption or inhibit other major intestinal transporters. Pharmacokinetics in mice showed >90% oral bioavailability with sustained therapeutic serum levels for >4 h without the significant toxicity seen with 7-d administration at 5 mg/kg/d. As evidence to support efficacy in human diarrheas, (R)-BPO-27 blocked fluid secretion in primary cultures of enteroids from human small intestine and anion current in enteroid monolayers. These studies support the potential utility of (R)-BPO-27 for therapy of CFTR-mediated secretory diarrheas.-Cil, O., Phuan, P.-W., Gillespie, A. M., Lee, S., Tradtrantip, L., Yin, J., Tse, M., Zachos, N. C., Lin, R., Donowitz, M., Verkman, A. S. Benzopyrimido-pyrrolo-oxazine-dione CFTR inhibitor (R)-BPO-27 for antisecretory therapy of diarrheas caused by bacterial enterotoxins.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Diarrhea/chemically induced , Diarrhea/drug therapy , Oxazines/pharmacology , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Humans , Intestines/drug effects , Mice , Molecular Structure , Oxazines/chemical synthesis , Pyrimidinones/chemical synthesis , Pyrroles/chemical synthesis
10.
J Biol Chem ; 291(8): 3759-66, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26677228

ABSTRACT

Identification of Lgr5 as the intestinal stem cell marker as well as the growth factors necessary to replicate adult intestinal stem cell division has led to the establishment of the methods to generate "indefinite" ex vivo primary intestinal epithelial cultures, termed "mini-intestines." Primary cultures developed from isolated intestinal crypts or stem cells (termed enteroids/colonoids) and from inducible pluripotent stem cells (termed intestinal organoids) are being applied to study human intestinal physiology and pathophysiology with great expectations for translational applications, including regenerative medicine. Here we discuss the physiologic properties of these cultures, their current use in understanding diarrhea-causing host-pathogen interactions, and potential future applications.


Subject(s)
Adult Stem Cells/metabolism , Antigens, Differentiation/metabolism , Diarrhea , Intestinal Mucosa , Intestines , Organoids , Receptors, G-Protein-Coupled/metabolism , Adult Stem Cells/pathology , Diarrhea/metabolism , Diarrhea/pathology , Diarrhea/physiopathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Intestinal Mucosa/metabolism , Intestines/pathology , Intestines/physiopathology , Organoids/metabolism , Organoids/pathology , Organoids/physiopathology
11.
Hum Mol Genet ; 24(23): 6614-23, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26358773

ABSTRACT

Congenital sodium diarrhea (CSD) refers to an intractable diarrhea of intrauterine onset with high fecal sodium loss. CSD is clinically and genetically heterogeneous. Syndromic CSD is caused by SPINT2 mutations. While we recently described four cases of the non-syndromic form of CSD that were caused by dominant activating mutations in intestinal receptor guanylate cyclase C (GC-C), the genetic cause for the majority of CSD is still unknown. Therefore, we aimed to determine the genetic cause for non-GC-C non-syndromic CSD in 18 patients from 16 unrelated families applying whole-exome sequencing and/or chromosomal microarray analyses and/or direct Sanger sequencing. SLC9A3 missense, splicing and truncation mutations, including an instance of uniparental disomy, and whole-gene deletion were identified in nine patients from eight families with CSD. Two of these nine patients developed inflammatory bowel disease (IBD) at 4 and 16 years of age. SLC9A3 encodes Na(+)/H(+) antiporter 3 (NHE3), which is the major intestinal brush-border Na(+)/H(+) exchanger. All mutations were in the NHE3 N-terminal transport domain, and all missense mutations were in the putative membrane-spanning domains. Identified SLC9A3 missense mutations were functionally characterized in plasma membrane NHE null fibroblasts. SLC9A3 missense mutations compromised NHE3 activity by reducing basal surface expression and/or loss of basal transport function of NHE3 molecules, whereas acute regulation was normal. This study identifies recessive mutations in NHE3, a downstream target of GC-C, as a cause of CSD and implies primary basal NHE3 malfunction as a predisposition for IBD in a subset of patients.


Subject(s)
Abnormalities, Multiple/genetics , Diarrhea/congenital , Metabolism, Inborn Errors/genetics , Mutation , Sodium-Hydrogen Exchangers/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Diarrhea/genetics , Diarrhea/metabolism , Diarrhea/physiopathology , Female , Genes, Recessive , Humans , Infant , Infant, Newborn , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/metabolism , Intestines/physiopathology , Male , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/physiopathology , Microvilli/metabolism , Oligonucleotide Array Sequence Analysis , Sodium-Hydrogen Exchanger 3 , Young Adult
12.
Gastroenterology ; 150(3): 638-649.e8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26677983

ABSTRACT

BACKGROUND & AIMS: Human intestinal crypt-derived enteroids are a model of intestinal ion transport that require validation by comparison with cell culture and animal models. We used human small intestinal enteroids to study neutral Na(+) absorption and stimulated fluid and anion secretion under basal and regulated conditions in undifferentiated and differentiated cultures to show their functional relevance to ion transport physiology and pathophysiology. METHODS: Human intestinal tissue specimens were obtained from an endoscopic biopsy or surgical resections performed at Johns Hopkins Hospital. Crypts were isolated, enteroids were propagated in culture, induced to undergo differentiation, and transduced with lentiviral vectors. Crypt markers, surface cell enzymes, and membrane ion transporters were characterized using quantitative reverse-transcription polymerase chain reaction, immunoblot, or immunofluorescence analyses. We used multiphoton and time-lapse confocal microscopy to monitor intracellular pH and luminal dilatation in enteroids under basal and regulated conditions. RESULTS: Enteroids differentiated upon withdrawal of WNT3A, yielding decreased crypt markers and increased villus-like characteristics. Na(+)/H(+) exchanger 3 activity was similar in undifferentiated and differentiated enteroids, and was affected by known inhibitors, second messengers, and bacterial enterotoxins. Forskolin-induced swelling was completely dependent on cystic fibrosis transmembrane conductance regulator and partially dependent on Na(+)/H(+) exchanger 3 and Na(+)/K(+)/2Cl(-) cotransporter 1 inhibition in undifferentiated and differentiated enteroids. Increases in cyclic adenosine monophosphate with forskolin caused enteroid intracellular acidification in HCO3(-)-free buffer. Cyclic adenosine monophosphate-induced enteroid intracellular pH acidification as part of duodenal HCO3(-) secretion appears to require cystic fibrosis transmembrane conductance regulator and electrogenic Na(+)/HCO3(-) cotransporter 1. CONCLUSIONS: Undifferentiated or crypt-like, and differentiated or villus-like, human enteroids represent distinct points along the crypt-villus axis; they can be used to characterize electrolyte transport processes along the vertical axis of the small intestine. The duodenal enteroid model showed that electrogenic Na(+)/HCO3(-) cotransporter 1 might be a target in the intestinal mucosa for treatment of secretory diarrheas.


Subject(s)
Cell Differentiation , Intestine, Small/metabolism , Membrane Transport Proteins/metabolism , Sodium/metabolism , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Intestinal Secretions/metabolism , Intestine, Small/pathology , Intestine, Small/physiopathology , Ion Transport , Kinetics , Membrane Transport Proteins/genetics , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Microscopy, Video , Time-Lapse Imaging , Tissue Culture Techniques , Transduction, Genetic , Transfection
13.
J Virol ; 90(1): 43-56, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26446608

ABSTRACT

UNLABELLED: Human gastrointestinal tract research is limited by the paucity of in vitro intestinal cell models that recapitulate the cellular diversity and complex functions of human physiology and disease pathology. Human intestinal enteroid (HIE) cultures contain multiple intestinal epithelial cell types that comprise the intestinal epithelium (enterocytes and goblet, enteroendocrine, and Paneth cells) and are physiologically active based on responses to agonists. We evaluated these nontransformed, three-dimensional HIE cultures as models for pathogenic infections in the small intestine by examining whether HIEs from different regions of the small intestine from different patients are susceptible to human rotavirus (HRV) infection. Little is known about HRVs, as they generally replicate poorly in transformed cell lines, and host range restriction prevents their replication in many animal models, whereas many animal rotaviruses (ARVs) exhibit a broader host range and replicate in mice. Using HRVs, including the Rotarix RV1 vaccine strain, and ARVs, we evaluated host susceptibility, virus production, and cellular responses of HIEs. HRVs infect at higher rates and grow to higher titers than do ARVs. HRVs infect differentiated enterocytes and enteroendocrine cells, and viroplasms and lipid droplets are induced. Heterogeneity in replication was seen in HIEs from different patients. HRV infection and RV enterotoxin treatment of HIEs caused physiological lumenal expansion detected by time-lapse microscopy, recapitulating one of the hallmarks of rotavirus-induced diarrhea. These results demonstrate that HIEs are a novel pathophysiological model that will allow the study of HRV biology, including host restriction, cell type restriction, and virus-induced fluid secretion. IMPORTANCE: Our research establishes HIEs as nontransformed cell culture models to understand human intestinal physiology and pathophysiology and the epithelial response, including host restriction of gastrointestinal infections such as HRV infection. HRVs remain a major worldwide cause of diarrhea-associated morbidity and mortality in children ≤5 years of age. Current in vitro models of rotavirus infection rely primarily on the use of animal rotaviruses because HRV growth is limited in most transformed cell lines and animal models. We demonstrate that HIEs are novel, cellularly diverse, and physiologically relevant epithelial cell cultures that recapitulate in vivo properties of HRV infection. HIEs will allow the study of HRV biology, including human host-pathogen and live, attenuated vaccine interactions; host and cell type restriction; virus-induced fluid secretion; cell-cell communication within the epithelium; and the epithelial response to infection in cultures from genetically diverse individuals. Finally, drug therapies to prevent/treat diarrheal disease can be tested in these physiologically active cultures.


Subject(s)
Intestine, Small/virology , Models, Theoretical , Organ Culture Techniques/methods , Rotavirus Infections/pathology , Rotavirus Infections/virology , Rotavirus/physiology , Virus Replication , Humans , Intestine, Small/physiology
14.
J Biol Chem ; 289(29): 20039-53, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24867958

ABSTRACT

NHERF1, NHERF2, and NHERF3 belong to the NHERF (Na(+)/H(+) exchanger regulatory factor) family of PSD-95/Discs-large/ZO-1 (PDZ) scaffolding proteins. Individually, each NHERF protein has been shown to be involved in the regulation of multiple receptors or transporters including Na(+)/H(+) exchanger 3 (NHE3). Although NHERF dimerizations have been reported, results have been inconsistent, and the physiological function of NHERF dimerizations is still unknown. The current study semiquantitatively compared the interaction strength among all possible homodimerizations and heterodimerizations of these three NHERF proteins by pulldown and co-immunoprecipitation assays. Both methods showed that NHERF2 and NHERF3 heterodimerize as the strongest interaction among all NHERF dimerizations. In vivo NHERF2/NHERF3 heterodimerization was confirmed by FRET and FRAP (fluorescence recovery after photobleach). NHERF2/NHERF3 heterodimerization is mediated by PDZ domains of NHERF2 and the C-terminal PDZ domain recognition motif of NHERF3. The NHERF3-4A mutant is defective in heterodimerization with NHERF2 and does not support the inhibition of NHE3 by carbachol. This suggests a role for NHERF2/NHERF3 heterodimerization in the regulation of NHE3 activity. In addition, both PDZ domains of NHERF2 could be simultaneously occupied by NHERF3 and another ligand such as NHE3, α-actinin-4, and PKCα, promoting formation of NHE3 macrocomplexes. This study suggests that NHERF2/NHERF3 heterodimerization mediates the formation of NHE3 macrocomplexes, which are required for the inhibition of NHE3 activity by carbachol.


Subject(s)
Carbachol/pharmacology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Substitution , Animals , Caco-2 Cells , Cell Line , Cricetinae , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutagenesis, Site-Directed , PDZ Domains , Phosphoproteins/genetics , Protein Multimerization , Rabbits , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics
15.
Am J Physiol Gastrointest Liver Physiol ; 308(8): G664-77, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25573173

ABSTRACT

Short bowel syndrome (SBS) is a devastating condition in which insufficient small intestinal surface area results in malnutrition and dependence on intravenous parenteral nutrition. There is an increasing incidence of SBS, particularly in premature babies and newborns with congenital intestinal anomalies. Tissue-engineered small intestine (TESI) offers a therapeutic alternative to the current standard treatment, intestinal transplantation, and has the potential to solve its biggest challenges, namely donor shortage and life-long immunosuppression. We have previously demonstrated that TESI can be generated from mouse and human small intestine and histologically replicates key components of native intestine. We hypothesized that TESI also recapitulates native small intestine function. Organoid units were generated from mouse or human donor intestine and implanted into genetically identical or immunodeficient host mice. After 4 wk, TESI was harvested and either fixed and paraffin embedded or immediately subjected to assays to illustrate function. We demonstrated that both mouse and human tissue-engineered small intestine grew into an appropriately polarized sphere of intact epithelium facing a lumen, contiguous with supporting mesenchyme, muscle, and stem/progenitor cells. The epithelium demonstrated major ultrastructural components, including tight junctions and microvilli, transporters, and functional brush-border and digestive enzymes. This study demonstrates that tissue-engineered small intestine possesses a well-differentiated epithelium with intact ion transporters/channels, functional brush-border enzymes, and similar ultrastructural components to native tissue, including progenitor cells, whether derived from mouse or human cells.


Subject(s)
Digestion , Intestinal Absorption , Intestinal Mucosa/physiology , Intestinal Mucosa/transplantation , Intestine, Small/physiology , Intestine, Small/transplantation , Tissue Engineering/methods , Animals , Aquaporins/metabolism , Biological Transport , Cell Differentiation , Cell Polarity , Cell Proliferation , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/physiology , Epithelial Cells/transplantation , Epithelial Cells/ultrastructure , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Organoids , Sodium-Hydrogen Exchangers/metabolism , Tight Junctions/physiology , Tight Junctions/ultrastructure , Time Factors , Tissue Culture Techniques
16.
J Biol Chem ; 288(23): 16960-16974, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23612977

ABSTRACT

Na(+)/H(+) exchanger regulatory factor (NHERF) proteins are a family of PSD-95/Discs-large/ZO-1 (PDZ)-scaffolding proteins, three of which (NHERFs 1-3) are localized to the brush border in kidney and intestinal epithelial cells. All NHERF proteins are involved in anchoring membrane proteins that contain PDZ recognition motifs to form multiprotein signaling complexes. In contrast to their predicted immobility, NHERF1, NHERF2, and NHERF3 were all shown by fluorescence recovery after photobleaching/confocal microscopy to be surprisingly mobile in the microvilli of the renal proximal tubule OK cell line. Their diffusion coefficients, although different among the three, were all of the same magnitude as that of the transmembrane proteins, suggesting they are all anchored in the microvilli but to different extents. NHERF3 moves faster than NHERF1, and NHERF2 moves the slowest. Several chimeras and mutants of NHERF1 and NHERF2 were made to determine which part of NHERF2 confers the slower mobility rate. Surprisingly, the slower mobility rate of NHERF2 was determined by a unique C-terminal domain, which includes a nonconserved region along with the ezrin, radixin, moesin (ERM) binding domain. Also, this C-terminal domain of NHERF2 determined its greater detergent insolubility and was necessary for the formation of larger multiprotein NHERF2 complexes. In addition, this NHERF2 domain was functionally significant in NHE3 regulation, being necessary for stimulation by lysophosphatidic acid of activity and increased mobility of NHE3, as well as necessary for inhibition of NHE3 activity by calcium ionophore 4-Br-A23187. Thus, multiple functions of NHERF2 require involvement of an additional domain in this protein.


Subject(s)
Cytoskeletal Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Caco-2 Cells , Calcimycin/analogs & derivatives , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Cytoskeletal Proteins/genetics , Humans , Kidney Tubules, Proximal/cytology , Lysophospholipids/pharmacology , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphoproteins/genetics , Protein Structure, Tertiary , Protein Transport/drug effects , Protein Transport/physiology , Rabbits , Rats , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics
17.
Clin Gastroenterol Hepatol ; 12(1): 27-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24184676

ABSTRACT

Diarrheal diseases remain a leading cause of morbidity and mortality for children in developing countries, while representing an important cause of morbidity worldwide. The World Health Organization recommended that low osmolarity oral rehydration solutions plus zinc save lives in patients with acute diarrhea, but there are no approved, safe drugs that have been shown to be effective against most causes of acute diarrhea. Identification of abnormalities in electrolyte handling by the intestine in diarrhea, including increased intestinal anion secretion and reduced Na(+) absorption, suggest a number of potential drug targets. This is based on the view that successful drug therapy for diarrhea will result from correcting the abnormalities in electrolyte transport that are pathophysiologic for diarrhea. We review the molecular mechanisms of physiologic regulation of intestinal ion transport and changes that occur in diarrhea and the status of drugs being developed to correct the transport abnormalities in Na(+) absorption that occur in diarrhea. Mechanisms of Cl(-) secretion and approaches to anti-Cl(-) secretory therapies of diarrhea are discussed in a companion review.


Subject(s)
Diarrhea/drug therapy , Electrolytes/metabolism , Sodium/metabolism , Water-Electrolyte Balance/drug effects , Humans , Molecular Medicine/trends
18.
Cell Physiol Biochem ; 33(3): 869-81, 2014.
Article in English | MEDLINE | ID: mdl-24713550

ABSTRACT

BACKGROUND: In intestinal epithelial cells, acute regulation of the brush border Na(+)/H(+) exchanger, NHE3, usually occurs by changes in endocytosis and/or exocytosis. Constitutive NHE3 endocytosis involves clathrin. Carbachol (CCH), which elevates intracellular Ca(2+) ([Ca(2+)]i), decreases NHE3 activity and stimulates endocytosis; however, the mechanism involved in calcium-mediated endocytosis of NHE3 is unclear. A pool of NHE3 resides in lipid rafts, which contributes to basal, but not cAMP-mediated, NHE3 trafficking, suggesting that an alternative mechanism exists for NHE3 endocytosis. Cdc42 was demonstrated to play an integral role in some cases of cholesterol-sensitive, clathrin-independent endocytosis. Therefore, the current study was designed to test the hypotheses that (1) clathrin-mediated endocytosis (CME) is involved in constitutive, but not CCH-mediated, endocytosis of NHE3, and (2) CCH-mediated endocytosis of NHE3 occurs through a lipid raft, activated Cdc42-dependent pathway that does not involve clathrin. METHODS: The role of Cdc42 and lipid rafts on NHE3 activity and endocytosis were investigated in polarized Caco-2/BBe cells using pharmacological and shRNA knockdown approaches. RESULTS: Basal NHE3 activity was increased in the presence of CME blockers (chlorpromazine; K(+) depletion) supporting previous reports that constitutive NHE3 endocytosis is clathrin dependent. In contrast, CCH-inhibition of NHE3 activity was abolished in Caco-2/BBe cells treated with MßCD (to disrupt lipid rafts) as well as in Cdc42 knockdown cells but was unaffected by CME blockers. CONCLUSION: CCH-mediated inhibition of NHE3 activity is not dependent on clathrin and involves lipid rafts and requires Cdc42.


Subject(s)
Carbachol/pharmacology , Clathrin/metabolism , Endocytosis/drug effects , Membrane Microdomains/metabolism , Sodium-Hydrogen Exchangers/metabolism , cdc42 GTP-Binding Protein/metabolism , Caco-2 Cells , Humans , Sodium-Hydrogen Exchanger 3
19.
Am J Physiol Cell Physiol ; 305(3): C266-75, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23703528

ABSTRACT

Elevated levels of intracellular Ca(2+) ([Ca(2+)]i) inhibit Na(+)/H(+) exchanger 3 (NHE3) activity in the intact intestine. We previously demonstrated that PLC-γ directly binds NHE3, an interaction that is necessary for [Ca(2+)]i inhibition of NHE3 activity, and that PLC-γ Src homology 2 (SH2) domains may scaffold Ca(2+) signaling proteins necessary for regulation of NHE3 activity. [Ca(2+)]i regulation of NHE3 activity is also c-Src dependent; however, the mechanism by which c-Src is involved is undetermined. We hypothesized that the SH2 domains of PLC-γ might link c-Src to NHE3-containing complexes to mediate [Ca(2+)]i inhibition of NHE3 activity. In Caco-2/BBe cells, carbachol (CCh) decreased NHE3 activity by ∼40%, an effect abolished with the c-Src inhibitor PP2. CCh treatment increased the amount of active c-Src as early as 1 min through increased Y(416) phosphorylation. Coimmunoprecipitation demonstrated that c-Src associated with PLC-γ, but not NHE3, under basal conditions, an interaction that increased rapidly after CCh treatment and occurred before the dissociation of PLC-γ and NHE3 that occurred 10 min after CCh treatment. Finally, direct binding to c-Src only occurred through the PLC-γ SH2 domains, an interaction that was prevented by blocking the PLC-γ SH2 domain. This study demonstrated that c-Src 1) activity is necessary for [Ca(2+)]i inhibition of NHE3 activity, 2) activation occurs rapidly (∼1 min) after CCh treatment, 3) directly binds PLC-γ SH2 domains and associates dynamically with PLC-γ under elevated [Ca(2+)]i conditions, and 4) does not directly bind NHE3. Under elevated [Ca(2+)]i conditions, PLC-γ scaffolds c-Src into NHE3-containing multiprotein complexes before dissociation of PLC-γ from NHE3 and subsequent endocytosis of NHE3.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Carbachol/pharmacology , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Caco-2 Cells , Calcium/chemistry , Calcium/metabolism , Cell Line, Tumor , Enzyme Activation , Genes, src , Humans , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins pp60(c-src)/metabolism , Pyrimidines/pharmacology , Signal Transduction , Sodium-Hydrogen Exchanger 3
20.
J Biol Chem ; 287(16): 13442-56, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22371496

ABSTRACT

The epithelial brush border (BB) Na(+)/H(+) exchanger 3 (NHE3) accounts for most renal and intestinal Na(+) absorption. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) inhibits NHE3 activity under basal conditions in intact intestine, acting in the BB, but the mechanism is unclear. We now demonstrate that in both PS120 fibroblasts and polarized Caco-2BBe cells expressing NHE3, CaMKII inhibits basal NHE3 activity, because the CaMKII-specific inhibitors KN-93 and KN-62 stimulate NHE3 activity. This inhibition requires NHERF2. CaMKIIγ associates with NHE3 between aa 586 and 605 in the NHE3 C terminus in a Ca(2+)-dependent manner, with less association when Ca(2+) is increased. CaMKII inhibits NHE3 by an effect on its turnover number, not changing surface expression. Back phosphorylation demonstrated that NHE3 is phosphorylated by CaMKII under basal conditions. This overall phosphorylation of NHE3 is not affected by the presence of NHERF2. Amino acids downstream of NHE3 aa 690 are required for CaMKII to inhibit basal NHE3 activity, and mutations of the three putative CaMKII phosphorylation sites downstream of aa 690 each prevented KN-93 stimulation of NHE3 activity. These studies demonstrate that CaMKIIγ is a novel NHE3-binding protein, and this association is reduced by elevated Ca(2+). CaMKII inhibits basal NHE3 activity associated with phosphorylation of NHE3 by effects requiring aa downstream of NHE3 aa 690 and of the CaMKII-binding site on NHE3. CaMKII binding to and phosphorylation of the NHE3 C terminus are parts of the physiologic regulation of NHE3 that occurs in fibroblasts as well as in the BB of an intestinal Na(+)-absorptive cell.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Microvilli/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Sodium-Hydrogen Exchangers/metabolism , Animals , Caco-2 Cells , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Epidermal Growth Factor/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mutagenesis/physiology , Phosphorylation/physiology , Protein Structure, Tertiary , Protons , Rabbits , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL