1
|
Kondrashina A, Arranz E, Cilla A, Faria MA, Santos-Hernández M, Miralles B, Hashemi N, Rasmussen MK, Young JF, Barberá R, Mamone G, Tomás-Cobos L, Bastiaan-Net S, Corredig M, Giblin L. Coupling in vitro food digestion with in vitro epithelial absorption; recommendations for biocompatibility. Crit Rev Food Sci Nutr 2024; 64:9618-9636. [PMID: 37233192 DOI: 10.1080/10408398.2023.2214628] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
As food transits the gastrointestinal tract, food structures are disrupted and nutrients are absorbed across the gut barrier. In the past decade, great efforts have focused on the creation of a consensus gastrointestinal digestion protocol (i.e., INFOGEST method) to mimic digestion in the upper gut. However, to better determine the fate of food components, it is also critical to mimic food absorption in vitro. This is usually performed by treating polarized epithelial cells (i.e., differentiated Caco-2 monolayers) with food digesta. This food digesta contains digestive enzymes and bile salts, and if following the INFOGEST protocol, at concentrations that although physiologically relevant are harmful to cells. The lack of a harmonized protocol on how to prepare the food digesta samples for downstream Caco-2 studies creates challenges in comparing inter laboratory results. This article aims to critically review the current detoxification practices, highlight potential routes and their limitations, and recommend common approaches to ensure food digesta is biocompatible with Caco-2 monolayers. Our ultimate aim is to agree a harmonized consensus protocol or framework for in vitro studies focused on the absorption of food components across the intestinal barrier.
Collapse
Affiliation(s)
- Alina Kondrashina
- Global Research and Technology Centre, H&H Group, H&H Research, Fermoy, Ireland
| | - Elena Arranz
- Department of Nutrition and Food Science, Faculty of Pharmacy, Complutense University of Madrid (UCM), Madrid, Spain
| | - Antonio Cilla
- Nutrition and Food Science Area, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Miguel A Faria
- LAQV/REQUIMTE, Laboratório de Bromatologia e Hidrologia, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, Porto, Portugal
| | - Marta Santos-Hernández
- Wellcome Trust - MRC Institute of Metabolic Science, Metabolic Research laboratories, Addenbrooke's Hospital, Cambridge, UK
| | - Beatriz Miralles
- Institute of Food Science Research CIAL (CSIC-UAM), Madrid, Spain
| | - Negin Hashemi
- Department of Food Science, Aarhus University, Agro Food Park 48, Aarhus, Denmark
| | | | - Jette F Young
- Department of Food Science, Aarhus University, Agro Food Park 48, Aarhus, Denmark
| | - Reyes Barberá
- Nutrition and Food Science Area, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Gianfranco Mamone
- Institute of Food Sciences - National Research Council, Avellino, Italy
| | - Lidia Tomás-Cobos
- In vitro preclinical studies department, AINIA, Avenida Benjamín Franklin 5-11, Parque Tecnológico de Valencia, Paterna, Spain
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, WG Wageningen, The Netherlands
| | - Milena Corredig
- Department of Food Science, Aarhus University, Agro Food Park 48, Aarhus, Denmark
| | - Linda Giblin
- Teagasc Food Research Centre, Moorepark, Fermoy, Co Cork, Ireland
| |
Collapse
|
2
|
McConnell BB, Liang Z, Xu C, Han Y, Yun CC. LPA 5-Dependent signaling regulates regeneration of the intestinal epithelium following irradiation. Am J Physiol Gastrointest Liver Physiol 2024; 326:G631-G642. [PMID: 38593468 PMCID: PMC11376986 DOI: 10.1152/ajpgi.00269.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/20/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid molecule that regulates a wide array of cellular functions, including proliferation, differentiation, and survival, via activation of cognate receptors. The LPA5 receptor is highly expressed in the intestinal epithelium, but its function in restoring intestinal epithelial integrity following injury has not been examined. Here, we use a radiation-induced injury model to study the role of LPA5 in regulating intestinal epithelial regeneration. Control mice (Lpar5f/f) and mice with an inducible, epithelial cell-specific deletion of Lpar5 in the small intestine (Lpar5IECKO) were subjected to 10 Gy total body X-ray irradiation and analyzed during recovery. Repair of the intestinal mucosa was delayed in Lpar5IECKO mice with reduced epithelial proliferation and increased crypt cell apoptosis. These effects were accompanied by reduced numbers of OLFM4+ intestinal stem cells (ISCs). The effects of LPA5 on ISCs were corroborated by studies using organoids derived from Lgr5-lineage tracking reporter mice with deletion of Lpar5 in Lgr5+-stem cells (Lgr5Cont or Lgr5ΔLpar5). Irradiation of organoids resulted in fewer numbers of Lgr5ΔLpar5 organoids retaining Lgr5+-derived progenitor cells compared with Lgr5Cont organoids. Finally, we observed that impaired regeneration in Lpar5IECKO mice was associated with reduced numbers of Paneth cells and decreased expression of Yes-associated protein (YAP), a critical factor for intestinal epithelial repair. Our study highlights a novel role for LPA5 in regeneration of the intestinal epithelium following irradiation and its effect on the maintenance of Paneth cells that support the stem cell niche.NEW & NOTEWORTHY We used mice lacking expression of the lysophosphatidic acid receptor 5 (LPA5) in intestinal epithelial cells and intestinal organoids to show that the LPA5 receptor protects intestinal stem cells and progenitors from radiation-induced injury. We show that LPA5 induces YAP signaling and regulates Paneth cells.
Collapse
Affiliation(s)
- Beth B McConnell
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Zhongxing Liang
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Chad Xu
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
3
|
Han Y, Bagchi P, Yun CC. Regulation of the intestinal Na +/H + exchanger NHE3 by AMP-activated kinase is dependent on phosphorylation of NHE3 at S555 and S563. Am J Physiol Cell Physiol 2024; 326:C50-C59. [PMID: 38047302 PMCID: PMC11192475 DOI: 10.1152/ajpcell.00540.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/05/2023]
Abstract
Electroneutral NaCl transport by Na+/H+ exchanger 3 (NHE3, SLC9A3) is the major Na+ absorptive mechanism in the intestine and decreased NHE3 activity contributes to diarrhea. Patients with diabetes often experience gastrointestinal adverse effects and medications are often a culprit for chronic diarrhea in type 2 diabetes (T2D). We have shown previously that metformin, the most widely prescribed drug for the treatment of T2D, induces diarrhea by inhibition of Na+/H+ exchanger 3 (NHE3) in rodent models of T2D. Metformin was shown to activate AMP-activated protein kinase (AMPK), but AMPK-independent glycemic effects of metformin are also known. The current study is undertaken to determine whether metformin inhibits NHE3 by activation of AMPK and the mechanism by which NHE3 is inhibited by AMPK. Inhibition of NHE3 by metformin was abolished by knockdown of AMPK-α1 or AMPK-α2. AMPK activation by 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) phosphorylated NHE3 at S555. S555 is the primary site of phosphorylation by protein kinase A (PKA), but AMPK phosphorylated S555 independently of PKA. Using Mass spectrometry, we found S563 as a newly recognized phosphorylation site in NHE3. Altering either S555 or S563 to Ala was sufficient to block the inhibition of NHE3 activity by AMPK. NHE3 inhibition is dependent on ubiquitination by the E3 ubiquitin ligase Nedd4-2 and metformin was shown to induce NHE3 internalization via Nedd4-2-mediated ubiquitination. AICAR did not increase NHE3 ubiquitination when S555 or S563 was mutated. We conclude that AMPK activation inhibits NHE3 activity and NHE3 inhibition is associated with phosphorylation of NHE3 at S555 and S563.NEW & NOTEWORTHY We show that AMP-activated protein kinase (AMPK) phosphorylates NHE3 at S555 and S563 to inhibit NHE3 activity in intestinal epithelial cells. Phosphorylation of NHE3 by AMPK is necessary for ubiquitination of NHE3.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University, Atlanta, Georgia, United States
| | - C Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
4
|
Han Y, Srinivasan S, Yun CC. Inhibition of protein kinase C-α and activation of ezrin by Lactobacillus acidophilus restore Na +/H + exchange activity and fluid absorption in db/db mice. Am J Physiol Endocrinol Metab 2023; 325:E214-E226. [PMID: 37467022 PMCID: PMC10511175 DOI: 10.1152/ajpendo.00145.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023]
Abstract
Gastrointestinal (GI) complications, including diarrhea, constipation, and gastroparesis, are common in patients with diabetes. Dysregulation of the Na+/H+ exchanger NHE3 in the intestine is linked to diarrhea and constipation, and recent studies showed that NHE3 expression is reduced in type 1 diabetes and metformin causes diarrhea in the db/db mouse model of type 2 diabetes (T2D) via inhibition of NHE3. In this study, we investigated whether NHE3 expression is altered in type 2 diabetic intestine and the underlying mechanism that dysregulates NHE3. NHE3 expression in the brush border membrane (BBM) of the intestine of diabetic mice and humans was decreased. Protein kinase C (PKC) activation is associated with pathologies of diabetes, and immunofluorescence (IF) analysis revealed increased BBM PKCα abundance. Inhibition of PKCα increased NHE3 BBM abundance and NHE3-mediated intestinal fluid absorption in db/db mice. Previous studies have shown that Lactobacillus acidophilus (LA) stimulates intestinal ion transporters. LA increased NHE3 BBM expression and mitigated metformin-mediated inhibition of NHE3 in vitro and in vivo. To understand the underlying mechanism of LA-mediated stimulation of NHE3, we used Caco-2bbe cells overexpressing PKCα that mimic the elevated state of PKCα in T2D. LA diminished PKCα BBM expression, increased phosphorylation of ezrin, and the interaction of NHE3 with NHE regulatory factor 2 (NHERF2). In addition, inhibition of PKCι blocked phosphorylation of ezrin and activation of NHE3 by LA. These findings demonstrate that NHE3 is downregulated in T2D, and LA restores NHE3 expression via regulation of PKCα, PKCι, and ezrin.NEW & NOTEWORTHY We used mouse models of type 2 diabetes (T2D) and human patient-derived samples to show that Na+/H+ exchanger 3 (NHE3) expression is decreased in T2D. We show that protein kinase C-α (PKCα) is activated in diabetes and inhibition of PKCα increased NHE3 expression and mitigates diarrhea. We show that Lactobacillus acidophilus (LA) stimulates NHE3 via inhibition of PKCα and phosphorylation of ezrin.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Shanthi Srinivasan
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - C Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, Georgia, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
5
|
Complexification of In Vitro Models of Intestinal Barriers, A True Challenge for a More Accurate Alternative Approach. Int J Mol Sci 2023; 24:ijms24043595. [PMID: 36835003 PMCID: PMC9958734 DOI: 10.3390/ijms24043595] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
The use of cell models is common to mimic cellular and molecular events in interaction with their environment. In the case of the gut, the existing models are of particular interest to evaluate food, toxicants, or drug effects on the mucosa. To have the most accurate model, cell diversity and the complexity of the interactions must be considered. Existing models range from single-cell cultures of absorptive cells to more complex combinations of two or more cell types. This work describes the existing solutions and the challenges that remain to be solved.
Collapse
|
6
|
Anbazhagan AN, Priyamvada S, Kumar A, Jayawardena D, Borthakur A, Gill RK, Alrefai WA, Dudeja PK, Saksena S. Downregulation of NHE-3 (SLC9A3) expression by MicroRNAs in intestinal epithelial cells. Am J Physiol Cell Physiol 2022; 323:C1720-C1727. [PMID: 36189974 PMCID: PMC9722255 DOI: 10.1152/ajpcell.00294.2022] [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: 07/06/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022]
Abstract
Na+/H+ exchanger-3 (NHE-3) is the major apical membrane transporter involved in vectorial Na+ absorption in the intestine. Dysregulation of NHE-3 expression and/or function has been implicated in pathophysiology of diarrhea associated with gut inflammation and infections. Therefore, it is critical to understand the mechanisms involved in the regulation of NHE-3 expression. MicroRNAs (miRNAs) are highly conserved small RNAs that can regulate gene expression at the posttranscriptional level. To date, however, very little is known about the regulation of NHE-3 expression by microRNAs. Therefore, current studies were undertaken to examine the potential miRNA candidates that can regulate the expression of NHE-3 in intestinal epithelial cells. In silico analysis, using different algorithms, predicted several miRNAs that target NHE-3. MicroRNAs with highest context and target score, miR-326, miR-744-5p, and miR-330-5p, were selected for the current study. Human NHE-3 gene 3' untranslated region [3'UTR; 160 base pair (bp)] was cloned into pmirGLO vector upstream of luciferase reporter and transiently transfected with mimics of miR-326, miR-744-5p, and miR-330-5p into Caco-2, HT-29, and SK-CO15 cells. Cotransfection of NHE-3 3' UTR with miR-326 and -miR-330-5p mimics resulted in a significant decrease in relative luciferase activity. Transfection of miR-326 and -330-5p mimics into SK-CO15 cells significantly decreased the NHE-3 protein expression, with no change in NHE-3 messenger ribonucleic acid (mRNA) levels. Our findings demonstrate a novel mechanism for posttranscriptional regulation of NHE-3 by miR-326 and -330-5p by translational repression. We speculate that miR-326 and -330-5p dependent pathways may be involved in modulating NHE-3 expression under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Arivarasu N Anbazhagan
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Shubha Priyamvada
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Anoop Kumar
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| | - Dulari Jayawardena
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Alip Borthakur
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Ravinder K Gill
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Waddah A Alrefai
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| | - Pradeep K Dudeja
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|
7
|
Laubitz D, Gurney MA, Midura-Kiela M, Clutter C, Besselsen DG, Chen H, Ghishan FK, Kiela PR. Decreased NHE3 expression in colon cancer is associated with DNA damage, increased inflammation and tumor growth. Sci Rep 2022; 12:14725. [PMID: 36042372 PMCID: PMC9427942 DOI: 10.1038/s41598-022-19091-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Dysregulation of intra- and extracellular pH in cancer contributes to extracellular matrix remodeling, favors cell migration, proliferation, and metastasis. Although the primary attention has been focused on the role of the ubiquitous Na+/H+ exchanger isoform NHE1, the role of NHE3, the predominant apical isoform in colonic surface epithelium in the pathogenesis of colon cancer has not been investigated. Here, we show that NHE3 mRNA expression is significantly reduced in colorectal cancer patients and that low NHE3 expression is associated with poorer survival. Deletion of NHE3 in ApcMin mice evaluated at 15 weeks of age (significant mortality was observed beyond this time) led to lower body weights, increased mucosal inflammation, increased colonic tumor numbers, evidence of enhanced DNA damage in tumor surface epithelium, and to significant alteration in the gut microbiota. In the absence of the inflammatory and microbial pressors, ca. 70% knockdown of NHE3 expression in SK-CO15 cells led to reduced intracellular pH, elevated apical pH, dramatic differences in their transcriptomic profile, increased susceptibility to DNA damage, increased proliferation, decreased apoptosis and reduced adhesion to extracellular matrix proteins. Our findings suggest that loss of NHE3 in the surface epithelium of colonic tumors has profound consequences for cancer progression and behavior.
Collapse
Affiliation(s)
- Daniel Laubitz
- Department of Pediatrics, Steele Children's Research Center, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ, 85724, USA
| | - Michael A Gurney
- Department of Pediatrics, Steele Children's Research Center, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ, 85724, USA
| | - Monica Midura-Kiela
- Department of Pediatrics, Steele Children's Research Center, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ, 85724, USA
| | - Christy Clutter
- Department of Pediatrics, Steele Children's Research Center, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ, 85724, USA
| | | | - Hao Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fayez K Ghishan
- Department of Pediatrics, Steele Children's Research Center, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ, 85724, USA
| | - Pawel R Kiela
- Department of Pediatrics, Steele Children's Research Center, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ, 85724, USA.
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
8
|
Han Y, Yun CC. Metformin Inhibits Na +/H + Exchanger NHE3 Resulting in Intestinal Water Loss. Front Physiol 2022; 13:867244. [PMID: 35444557 PMCID: PMC9014215 DOI: 10.3389/fphys.2022.867244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 11/14/2022] Open
Abstract
Glycemic control is the key to the management of type 2 diabetes. Metformin is an effective, widely used drug for controlling plasma glucose levels in diabetes, but it is often the culprit of gastrointestinal adverse effects such as abdominal pain, nausea, indigestion, vomiting, and diarrhea. Diarrhea is a complex disease and altered intestinal transport of electrolytes and fluid is a common cause of diarrhea. Na+/H+ exchanger 3 (NHE3, SLC9A3) is the major Na+ absorptive mechanism in the intestine and our previous study has demonstrated that decreased NHE3 contributes to diarrhea associated with type 1 diabetes. The goal of this study is to investigate whether metformin regulates NHE3 and inhibition of NHE3 contributes to metformin-induced diarrhea. We first determined whether metformin alters intestinal water loss, the hallmark of diarrhea, in type 2 diabetic db/db mice. We found that metformin decreased intestinal water absorption mediated by NHE3. Metformin increased fecal water content although mice did not develop watery diarrhea. To determine the mechanism of metformin-mediated regulation of NHE3, we used intestinal epithelial cells. Metformin inhibited NHE3 activity and the effect of metformin on NHE3 was mimicked by a 5'-AMP-activated protein kinase (AMPK) activator and blocked by pharmacological inhibition of AMPK. Metformin increased phosphorylation and ubiquitination of NHE3, resulting in retrieval of NHE3 from the plasma membrane. Previous studies have demonstrated the role of neural precursor cell expressed, developmentally down-regulated 4-2 (Nedd4-2) in regulation of human NHE3. Silencing of Nedd4-2 mitigated NHE3 inhibition and ubiquitination by metformin. Our findings suggest that metformin-induced diarrhea in type 2 diabetes is in part caused by reduced Na+ and water absorption that is associated with NHE3 inhibition, probably by AMPK.
Collapse
Affiliation(s)
- Yiran Han
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - C. Chris Yun
- Gastroenterology Research, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
9
|
Jenkin KA, Han Y, Lin S, He P, Yun CC. Nedd4-2-dependent Ubiquitination Potentiates the Inhibition of Human NHE3 by Cholera Toxin and Enteropathogenic Escherichia coli. Cell Mol Gastroenterol Hepatol 2021; 13:695-716. [PMID: 34823064 PMCID: PMC8789535 DOI: 10.1016/j.jcmgh.2021.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Diarrhea is one of the most common illnesses and is often caused by bacterial infection. Recently, we have shown that human Na+/H+ exchanger NHE3 (hNHE3), but not non-human NHE3s, interacts with the E3 ubiquitin ligase Nedd4-2. We hypothesize that this property of hNHE3 contributes to the increased severity of diarrhea in humans. METHODS We used humanized mice expressing hNHE3 in the intestine (hNHE3int) to compare the contribution of hNHE3 and mouse NHE3 to diarrhea induced by cholera toxin (CTX) and enteropathogenic Escherichia coli (EPEC). We measured Na+/H+ exchange activity and fluid absorption. The role of Nedd4-2 on hNHE3 activity and ubiquitination was determined by knockdown in Caco-2bbe cells. The effects of protein kinase A (PKA), the primary mediator of CTX-induced diarrhea, on Nedd4-2 and hNHE3 phosphorylation and their interaction were determined. RESULTS The effects of CTX and EPEC were greater in hNHE3int mice than in control wild-type (WT) mice, resulting in greater inhibition of NHE3 activity and increased fluid accumulation in the intestine, the hallmark of diarrhea. Activation of PKA increased ubiquitination of hNHE3 and enhanced interaction of Nedd4-2 with hNHE3 via phosphorylation of Nedd4-2 at S342. S342A mutation mitigated the Nedd4-2-hNHE3 interaction and blocked PKA-induced inhibition of hNHE3. Unlike non-human NHE3s, inhibition of hNHE3 by PKA is independent of NHE3 phosphorylation, suggesting a distinct mechanism of hNHE3 regulation. CONCLUSIONS The effects of CTX and EPEC on hNHE3 are amplified, and the unique properties of hNHE3 may contribute to diarrheal symptoms occurring in humans.
Collapse
Affiliation(s)
- Kayte A. Jenkin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Songbai Lin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - C. Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia,Correspondence Address correspondence to: Chris Yun, PhD, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia 30324. fax: (404) 727-5767.
| |
Collapse
|
10
|
Control of Intestinal Epithelial Permeability by Lysophosphatidic Acid Receptor 5. Cell Mol Gastroenterol Hepatol 2021; 12:1073-1092. [PMID: 33975030 PMCID: PMC8350072 DOI: 10.1016/j.jcmgh.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Epithelial cells form a monolayer at mucosal surface that functions as a highly selective barrier. Lysophosphatidic acid (LPA) is a bioactive lipid that elicits a broad range of biological effects via cognate G protein-coupled receptors. LPA receptor 5 (LPA5) is highly expressed in intestinal epithelial cells, but its role in the intestine is not well-known. Here we determined the role of LPA5 in regulation of intestinal epithelial barrier. METHODS Epithelial barrier integrity was determined in mice with intestinal epithelial cell (IEC)-specific LPA5 deletion, Lpar5ΔIEC. LPA was orally administered to mice, and intestinal permeability was measured. Dextran sulfate sodium (DSS) was used to induce colitis. Human colonic epithelial cell lines were used to determine the LPA5-mediated signaling pathways that regulate epithelial barrier. RESULTS We observed increased epithelial permeability in Lpar5ΔIEC mice with reduced claudin-4 expression. Oral administration of LPA decreased intestinal permeability in wild-type mice, but the effect was greatly mitigated in Lpar5ΔIEC mice. Serum lipopolysaccharide level and bacterial loads in the intestine and liver were elevated in Lpar5ΔIEC mice. Lpar5ΔIEC mice developed more severe colitis induced with DSS. LPA5 transcriptionally regulated claudin-4, and this regulation was dependent on transactivation of the epidermal growth factor receptor, which induced localization of Rac1 at the cell membrane. LPA induced the translocation of Stat3 to the cell membrane and promoted the interaction between Rac1 and Stat3. Inhibition of Stat3 ablated LPA-mediated regulation of claudin-4. CONCLUSIONS This study identifies LPA5 as a regulator of the intestinal barrier. LPA5 promotes claudin-4 expression in IECs through activation of Rac1 and Stat3.
Collapse
|
11
|
Han Y, Yun CC. Ubiquitin-specific peptidase 7 (USP7) and USP10 mediate deubiquitination of human NHE3 regulating its expression and activity. FASEB J 2020; 34:16476-16488. [PMID: 33095475 DOI: 10.1096/fj.202001875r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/21/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Na+ /H+ exchanger NHE3 of human or primates differs from NHE3 of other animals by having a PY motif, which mediates interaction with the E3 ubiquitin (Ub) ligase Nedd4-2. Ub-conjugation of human NHE3 by Nedd4-2 regulates endocytosis of NHE3 but does not affect its cellular expression. Because Ub-conjugation is a reversible process, the aim of this study is to identify deubiquitinating enzyme (DUB) regulating the post-endosomal fate of human NHE3. Using an activity-based chemical screening, we identified ubiquitin specific protease-7 (USP7) and USP10 that bind NHE3. The roles of DUBs in regulation of NHE3 were studied by determining the effects of silencing of USP7 and USP10. Knockdown of USP7 or USP10 resulted in increased NHE3 ubiquitination and decreased NHE3 expression at the surface membrane and cellular level. The endocytic retrieval of NHE3 was promoted by depletion of USP7 or USP10, with increased association of NHE3 with Rab5a and Rab7. Inhibition of USP7 and USP10 by chemical inhibitors or knockdown had an additive effect on NHE3. In addition, NHE3 half-life was reduced accounting for decreased NHE3 protein abundance. NHE3 is inhibited by protein kinase A. Activation of PKA by forskolin decreased the binding of USP7 and USP10 to NHE3, while increasing ubiquitination of NHE3. Knockdown of USP10 had an additive effect on PKA-dependent inhibition of NHE3. These findings demonstrate that USP7 and USP10 are DUBs that regulate NHE3 ubiquitination and expression, and reveal a new mechanism of NHE3 inhibition involving DUBs.
Collapse
Affiliation(s)
- Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Gastroenterology Research, Atlanta VA Medical Center, Decatur, GA, USA.,Cell and Molecular Biology Research Program, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
12
|
Schoultz I, Keita ÅV. The Intestinal Barrier and Current Techniques for the Assessment of Gut Permeability. Cells 2020; 9:E1909. [PMID: 32824536 PMCID: PMC7463717 DOI: 10.3390/cells9081909] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/08/2023] Open
Abstract
The intestinal barrier is essential in human health and constitutes the interface between the outside and the internal milieu of the body. A functional intestinal barrier allows absorption of nutrients and fluids but simultaneously prevents harmful substances like toxins and bacteria from crossing the intestinal epithelium and reaching the body. An altered intestinal permeability, a sign of a perturbed barrier function, has during the last decade been associated with several chronic conditions, including diseases originating in the gastrointestinal tract but also diseases such as Alzheimer and Parkinson disease. This has led to an intensified interest from researchers with diverse backgrounds to perform functional studies of the intestinal barrier in different conditions. Intestinal permeability is defined as the passage of a solute through a simple membrane and can be measured by recording the passage of permeability markers over the epithelium via the paracellular or the transcellular route. The methodological tools to investigate the gut barrier function are rapidly expanding and new methodological approaches are being developed. Here we outline and discuss, in vivo, in vitro and ex vivo techniques and how these methods can be utilized for thorough investigation of the intestinal barrier.
Collapse
Affiliation(s)
- Ida Schoultz
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, 703 62 Örebro, Sweden;
| | - Åsa V. Keita
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
13
|
Enteropathogenic Escherichia coli (EPEC) Recruitment of PAR Polarity Protein Atypical PKCζ to Pedestals and Cell-Cell Contacts Precedes Disruption of Tight Junctions in Intestinal Epithelial Cells. Int J Mol Sci 2020; 21:ijms21020527. [PMID: 31947656 PMCID: PMC7014222 DOI: 10.3390/ijms21020527] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/26/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) uses a type three secretion system to inject effector proteins into host intestinal epithelial cells, causing diarrhea. EPEC induces the formation of pedestals underlying attached bacteria, disrupts tight junction (TJ) structure and function, and alters apico-basal polarity by redistributing the polarity proteins Crb3 and Pals1, although the mechanisms are unknown. Here we investigate the temporal relationship of PAR polarity complex and TJ disruption following EPEC infection. EPEC recruits active aPKCζ, a PAR polarity protein, to actin within pedestals and at the plasma membrane prior to disrupting TJ. The EPEC effector EspF binds the endocytic protein sorting nexin 9 (SNX9). This interaction impacts actin pedestal organization, recruitment of active aPKCζ to actin at cell–cell borders, endocytosis of JAM-A S285 and occludin, and TJ barrier function. Collectively, data presented herein support the hypothesis that EPEC-induced perturbation of TJ is a downstream effect of disruption of the PAR complex and that EspF binding to SNX9 contributes to this phenotype. aPKCζ phosphorylates polarity and TJ proteins and participates in actin dynamics. Therefore, the early recruitment of aPKCζ to EPEC pedestals and increased interaction with actin at the membrane may destabilize polarity complexes ultimately resulting in perturbation of TJ.
Collapse
|
14
|
Kumar A, Jayawardena D, Anbazhagan AN, Chatterjee I, Priyamvada S, Alrefai WA, Borthakur A, Dudeja PK. Decreased SLC26A3 expression and function in intestinal epithelial cells in response to Cryptosporidium parvum infection. Am J Physiol Cell Physiol 2019; 317:C1205-C1212. [PMID: 31483700 DOI: 10.1152/ajpcell.00278.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The protozoan parasite Cryptosporidium parvum (CP) causes cryptosporidiosis, a diarrheal disease worldwide. Infection in immunocompetent hosts typically results in acute, self-limiting, or recurrent diarrhea. However, in immunocompromised individuals infection can cause fulminant diarrhea, extraintestinal manifestations, and death. To date, the mechanisms underlying CP-induced diarrheal pathogenesis are poorly understood. Diarrheal diseases most commonly involve increased secretion and/or decreased absorption of fluid and electrolytes. We and others have previously shown impaired chloride absorption in infectious diarrhea due to dysregulation of SLC26A3 [downregulated in adenoma (DRA)], the human intestinal apical membrane Cl-/HCO3- exchanger protein. However, there are no studies on the effects of CP infection on DRA activity. Therefore, we examined the expression and function of DRA in intestinal epithelial cells in response to CP infection in vitro and in vivo. CP infection (0.5 × 106 oocysts/well in 24-well plates, 24 h) of Caco-2 cell monolayers significantly decreased Cl-/HCO3- exchange activity (measured as DIDS-sensitive 125I uptake) as well as DRA mRNA and protein levels. Substantial downregulation of DRA mRNA and protein was also observed following CP infection ex vivo in mouse enteroid-derived monolayers and in vivo in the ileal and jejunal mucosa of C57BL/6 mice for 24 h. However, at 48 h after infection in vivo, the effects on DRA mRNA and protein were attenuated and at 5 days after infection DRA returned to normal levels. Our results suggest that impaired chloride absorption due to downregulation of DRA could be one of the contributing factors to CP-induced acute, self-limiting diarrhea in immunocompetent hosts.
Collapse
Affiliation(s)
- Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Dulari Jayawardena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Arivarasu N Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Ishita Chatterjee
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Shubha Priyamvada
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Waddah A Alrefai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Alip Borthakur
- Department of Clinical and Translational Sciences, Marshall University Joan C. Edwards School of Medicine, Huntington, West Virginia
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois.,Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
15
|
Yu Y, Seidler A, Zhou K, Yuan Z, Yeruva S, Amiri M, Yun CC, Nikolovska K, Seidler U. Expression, Localization and Functional Activity of the Major Na⁺/H⁺ Exchange Isoforms Expressed in the Intestinal Cell Line Caco-2BBe. Cell Physiol Biochem 2019; 52:1017-1038. [PMID: 30977986 PMCID: PMC7104346 DOI: 10.33594/000000070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
Background/Aims Enterocytes express a number of NHE isoforms with presumed localization in the apical (NHE2, 3 and 8) or basolateral (NHE1) membrane. Functional activity and localization of enterocyte NHE isoforms were assessed using fully differentiated Caco-2BBe cells, whose genetic expression profile closely resembles mature enterocytes. Methods The activity of the different NHEs was analyzed by fluorometric pHi-metry in a perfusion chamber with separate apical and basolateral perfusion, using specific inhibitors and shRNA knockdown of NHE2. The expression of the NHEs and of other relevant acid extrusion transporters was quantified by qPCR. Results Quantitative comparison of the mRNA expression levels of the different NHE isoforms in 14 day-differentiated Caco-2BBe cells showed the following order: NHE2>NHE8>NHE3>NHE1. Acid-activated NHE exchange rates in the basolateral membrane were >6-fold higher than in the apical membrane. 79 ± 3 % of the acid-activated basolateral Na+/H+ exchange rate displayed a NHE1-typical inhibitor profile, and no NHE2/3/8 typical activity could be observed. Analysis of the apical Na+/H+ exchange rates revealed that approximately 51 ± 3 % of the total apical activity displayed a NHE2/8-typical inhibitor profile and 31 ± 6 % a NHE3-typical inhibitor profile. Because no selective NHE2 inhibitor is available, a stable NHE2 knockdown cell line (C2NHE2KD) was generated. C2NHE2KD displayed a reduced NHE2-typical apical Na+/H+ exchange rate and maintained a lower steady-state pHi, despite high expression levels of other acid extruders, in particular NBCn1 (Slc4a7). Conclusion Differentiated Caco-2BBe cells display particularly high mRNA expression levels of NHE2, which can be functionally identified in the apical membrane. Although at low intracellular pH, NHE2 transport rate was far lower than that of NHE1. NHE2 activity was nevertheless essential for the maintenance of the steady-state pHi of these cells.
Collapse
Affiliation(s)
- Yan Yu
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Anna Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Kunyan Zhou
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Zhenglin Yuan
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Sunil Yeruva
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Chris C Yun
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | | | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany,
| |
Collapse
|
16
|
Jenkin KA, He P, Yun CC. Expression of lysophosphatidic acid receptor 5 is necessary for the regulation of intestinal Na +/H + exchanger 3 by lysophosphatidic acid in vivo. Am J Physiol Gastrointest Liver Physiol 2018; 315:G433-G442. [PMID: 29792531 PMCID: PMC6230692 DOI: 10.1152/ajpgi.00130.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid molecule, which regulates a broad range of pathophysiological processes. Recent studies have demonstrated that LPA modulates electrolyte flux in the intestine, and its potential as an antidiarrheal agent has been suggested. Of six LPA receptors, LPA5 is highly expressed in the intestine. Recent studies by our group have demonstrated activation of Na+/H+ exchanger 3 (NHE3) by LPA5. However, much of what has been elucidated was achieved using colonic cell lines that were transfected to express LPA5. In the current study, we engineered a mouse that lacks LPA5 in intestinal epithelial cells, Lpar5ΔIEC, and investigated the role of LPA5 in NHE3 regulation and fluid absorption in vivo. The intestine of Lpar5ΔIEC mice appeared morphologically normal, and the stool frequency and fecal water content were unchanged compared with wild-type mice. Basal rates of NHE3 activity and fluid absorption and total NHE3 expression were not changed in Lpar5ΔIEC mice. However, LPA did not activate NHE3 activity or fluid absorption in Lpar5ΔIEC mice, providing direct evidence for the regulatory role of LPA5. NHE3 activation involves trafficking of NHE3 from the terminal web to microvilli, and this mobilization of NHE3 by LPA was abolished in Lpar5ΔIEC mice. Dysregulation of NHE3 was specific to LPA, and insulin and cholera toxin were able to stimulate and inhibit NHE3, respectively, in both wild-type and Lpar5ΔIEC mice. The current study for the first time demonstrates the necessity of LPA5 in LPA-mediated stimulation of NHE3 in vivo. NEW & NOTEWORTHY This study is the first to assess the role of LPA5 in NHE3 regulation and fluid absorption in vivo using a mouse that lacks LPA5 in intestinal epithelial cells, Lpar5ΔIEC. Basal rates of NHE3 activity and fluid absorption, and total NHE3 expression were not changed in Lpar5ΔIEC mice. However, LPA did not activate NHE3 activity or fluid absorption in Lpar5ΔIEC mice, providing direct evidence for the regulatory role of LPA5.
Collapse
Affiliation(s)
- Kayte A. Jenkin
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Peijian He
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - C. Chris Yun
- 1Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,2Atlanta Veterans’ Affairs Medical Center, Decatur, Georgia,3Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
17
|
Romero-Calvo I, Ocón B, Gámez-Belmonte R, Hernández-Chirlaque C, de Jonge HR, Bijvelds MJ, Martínez-Augustin O, Sánchez de Medina F. Adenylyl cyclase 6 is involved in the hyposecretory status of experimental colitis. Pflugers Arch 2018; 470:1705-1717. [PMID: 30094477 DOI: 10.1007/s00424-018-2187-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/12/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022]
Abstract
One of the cardinal symptoms of intestinal inflammation is diarrhea. Acute intestinal inflammation is associated with inhibition of ion absorption and increased secretion, along with fluid leakage due to epithelial injury and changes in permeability. However, in the chronic situation, a downregulation of both absorptive and secretory transport has been reported. We investigated how experimental colitis reduces cAMP levels in intestinal epithelial cells through modulation of adenylyl cyclases (AC). Primary colonic epithelial cells obtained from rats with trinitrobenzenesulfonic acid colitis and non-colitic controls were analyzed for AC expression by RT-qPCR and Western blot, following a preliminary microarray analysis. AC6 and AC5 were found to be expressed in colonocytes, and downregulated by inflammation, with the former exhibiting considerably higher mRNA levels in both cases. To test the hypothesis that inflammatory cytokines may account for this effect, Caco 2 cells were treated with IL-1β, TNF-α, or IFN-γ. All three cytokines inhibited forskolin evoked short-circuit currents in Ussing chambers and lowered intracellular cAMP, but failed to alter AC6 mRNA levels. AC5/AC6 expression was however inhibited in mouse jejunal organoids treated with IFN-γ and TNF-α, but not IL-1β. Gene knockdown of AC6 resulted in a significant decrease of ion secretion in T84 cells. We conclude that the disturbances in ion secretion observed in rat TNBS colitis are associated with low intracellular levels of cAMP in the epithelium, which may be explained in part by the downregulation of AC5/AC6 expression by proinflammatory cytokines.
Collapse
Affiliation(s)
- Isabel Romero-Calvo
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Borja Ocón
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Reyes Gámez-Belmonte
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| | - Cristina Hernández-Chirlaque
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071, Granada, Spain
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Marcel J Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Campus de Cartuja s/n, 18071, Granada, Spain.
| | - Fermín Sánchez de Medina
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain
| |
Collapse
|
18
|
Kumar A, Malhotra P, Coffing H, Priyamvada S, Anbazhagan AN, Krishnan HR, Gill RK, Alrefai WA, Gavin DP, Pandey SC, Dudeja PK, Saksena S. Epigenetic modulation of intestinal Na +/H + exchanger-3 expression. Am J Physiol Gastrointest Liver Physiol 2018; 314:G309-G318. [PMID: 29167115 PMCID: PMC5899241 DOI: 10.1152/ajpgi.00293.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/01/2017] [Accepted: 11/09/2017] [Indexed: 01/31/2023]
Abstract
Na+/H+ exchanger-3 (NHE3) is crucial for intestinal Na+ absorption, and its reduction has been implicated in infectious and inflammatory bowel diseases (IBD)-associated diarrhea. Epigenetic mechanisms such as DNA methylation are involved in the pathophysiology of IBD. Whether changes in DNA methylation are involved in modulating intestinal NHE3 gene expression is not known. Caco-2 and HuTu 80 cells were used as models of human intestinal epithelial cells. Normal C57/BL6, wild-type, or growth arrest and DNA damage-inducible 45b (GADD45b) knockout (KO) mice were used as in vivo models. NHE3 gene DNA methylation levels were assessed by MBDCap (MethyMiner) assays. Results demonstrated that in vitro methylation of NHE3 promoter construct (p-1509/+127) cloned into a cytosine guanine dinucleotide-free lucia vector decreased the promoter activity in Caco-2 cells. DNA methyltransferase inhibitor 5-azacytidine (10 μM, 24 h) caused a significant decrease in DNA methylation of the NHE3 gene and concomitantly increased NHE3 expression in Caco-2 cells. Similarly, 5-azacytidine treatment increased NHE3 mRNA levels in HuTu 80 cells. 5-Azacytidine treatment for 3 wk (10 mg/kg body wt ip, 3 times/wk) also resulted in an increase in NHE3 expression in the mouse ileum and colon. Small-interfering RNA knockdown of GADD45b (protein involved in DNA demethylation) in Caco-2 cells decreased NHE3 mRNA expression. Furthermore, there was a significant decrease in NHE3 mRNA and protein expression in the ileum and colon of GADD45b KO mice. Our findings demonstrate that NHE3 gene expression is regulated by changes in its DNA methylation. NEW & NOTEWORTHY Our studies for the first time demonstrate that Na+/H+ exchanger-3 gene expression is regulated by an epigenetic mechanism involving DNA methylation.
Collapse
Affiliation(s)
- Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Pooja Malhotra
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Hayley Coffing
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Shubha Priyamvada
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Arivarasu N Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Harish R Krishnan
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago , Chicago, Illinois
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Waddah A Alrefai
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| | - David P Gavin
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago , Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago , Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
| |
Collapse
|
19
|
Vogel GF, van Rijn JM, Krainer IM, Janecke AR, Posovszky C, Cohen M, Searle C, Jantchou P, Escher JC, Patey N, Cutz E, Müller T, Middendorp S, Hess MW, Huber LA. Disrupted apical exocytosis of cargo vesicles causes enteropathy in FHL5 patients with Munc18-2 mutations. JCI Insight 2017; 2:94564. [PMID: 28724787 DOI: 10.1172/jci.insight.94564] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/06/2017] [Indexed: 01/07/2023] Open
Abstract
Familial hemophagocytic lymphohistiocytosis 5 (FHL5) is an autosomal recessive disease caused by mutations in STXBP2, coding for Munc18-2, which is required for SNARE-mediated membrane fusion. FHL5 causes hematologic and gastrointestinal symptoms characterized by chronic enteropathy that is reminiscent of microvillus inclusion disease (MVID). However, the molecular pathophysiology of FHL5-associated diarrhea is poorly understood. Five FHL5 patients, including four previously unreported patients, were studied. Morphology of duodenal sections was analyzed by electron and fluorescence microscopy. Small intestinal enterocytes and organoid-derived monolayers displayed the subcellular characteristics of MVID. For the analyses of Munc18-2-dependent SNARE-protein interactions, a Munc18-2 CaCo2-KO model cell line was generated by applying CRISPR/Cas9 technology. Munc18-2 is required for Slp4a/Stx3 interaction in fusion of cargo vesicles with the apical plasma membrane. Cargo trafficking was investigated in patient biopsies, patient-derived organoids, and the genome-edited model cell line. Loss of Munc18-2 selectively disrupts trafficking of certain apical brush-border proteins (NHE3 and GLUT5), while transport of DPPIV remained unaffected. Here, we describe the molecular mechanism how the loss of function of Munc18-2 leads to cargo-selective mislocalization of brush-border components and a subapical accumulation of cargo vesicles, as it is known from the loss of polarity phenotype in MVID.
Collapse
Affiliation(s)
- Georg F Vogel
- Department of Paediatrics I and.,Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jorik M van Rijn
- Division of Paediatrics, Department of Paediatric Gastroenterology and Regenerative Medicine Center Utrecht, Wilhelmina Children's Hospital, University Medical Centre (UMC) Utrecht, Utrecht, The Netherlands
| | - Iris M Krainer
- Department of Paediatrics I and.,Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Carsten Posovszky
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Marta Cohen
- Sheffield Children's Hospital NHS Trust, Western Bank, Sheffield, United Kingdom
| | - Claire Searle
- Clinical Genetics, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Prevost Jantchou
- Gastroentérologie Hépatologie et Nutrition Pédiatrique Hôpital Sainte-Justine, Université de Montréal, Montréal, Quebec, Canada
| | - Johanna C Escher
- Department of Pediatric Gastroenterology, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands
| | - Natalie Patey
- Clinical Genetics, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Ernest Cutz
- The Hospital for Sick Children, Toronto, Canada
| | | | - Sabine Middendorp
- Division of Paediatrics, Department of Paediatric Gastroenterology and Regenerative Medicine Center Utrecht, Wilhelmina Children's Hospital, University Medical Centre (UMC) Utrecht, Utrecht, The Netherlands
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
Espejo AB, Gao G, Black K, Gayatri S, Veland N, Kim J, Chen T, Sudol M, Walker C, Bedford MT. PRMT5 C-terminal Phosphorylation Modulates a 14-3-3/PDZ Interaction Switch. J Biol Chem 2017; 292:2255-2265. [PMID: 28031468 PMCID: PMC5313098 DOI: 10.1074/jbc.m116.760330] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 12/21/2016] [Indexed: 11/06/2022] Open
Abstract
PRMT5 is the primary enzyme responsible for the deposition of the symmetric dimethylarginine in mammalian cells. In an effort to understand how PRMT5 is regulated, we identified a threonine phosphorylation site within a C-terminal tail motif, which is targeted by the Akt/serum- and glucocorticoid-inducible kinases. While investigating the function of this posttranslational modification, we serendipitously discovered that its free C-terminal tail binds PDZ domains (when unphosphorylated) and 14-3-3 proteins (when phosphorylated). In essence, a phosphorylation event within the last few residues of the C-terminal tail generates a posttranslational modification-dependent PDZ/14-3-3 interaction "switch." The C-terminal motif of PRMT5 is required for plasma membrane association, and loss of this switching capacity is not compatible with life. This signaling phenomenon was recently reported for the HPV E6 oncoprotein but has not yet been observed for mammalian proteins. To investigate the prevalence of PDZ/14-3-3 switching in signal transduction, we built a protein domain microarray that harbors PDZ domains and 14-3-3 proteins. We have used this microarray to interrogate the C-terminal tails of a small group of candidate proteins and identified ERBB4, PGHS2, and IRK1 (as well as E6 and PRMT5) as conforming to this signaling mode, suggesting that PDZ/14-3-3 switching may be a broad biological paradigm.
Collapse
Affiliation(s)
- Alexsandra B Espejo
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
- the University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030
| | - Guozhen Gao
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
| | - Karynne Black
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
| | - Sitaram Gayatri
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
- the University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030
| | - Nicolas Veland
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
- the University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030
| | - Jeesun Kim
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
| | - Taiping Chen
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957
| | - Marius Sudol
- the Department of Physiology, National University of Singapore, Mechanobiology Institute and Institute for Molecular and Cell Biology (IMCB, A*STAR), Singapore 117597, Singapore, and
| | - Cheryl Walker
- the Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030
| | - Mark T Bedford
- From the Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957,
| |
Collapse
|
21
|
Deevi RK, McClements J, McCloskey KD, Fatehullah A, Tkocz D, Javadi A, Higginson R, Durban VM, Jansen M, Clarke A, Loughrey MB, Campbell FC. Vitamin D3 suppresses morphological evolution of the cribriform cancerous phenotype. Oncotarget 2016; 7:49042-49064. [PMID: 27119498 PMCID: PMC5226489 DOI: 10.18632/oncotarget.8863] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/28/2016] [Indexed: 12/14/2022] Open
Abstract
Development of cribriform morphology (CM) heralds malignant change in human colon but lack of mechanistic understanding hampers preventive therapy. This study investigated CM pathobiology in three-dimensional (3D) Caco-2 culture models of colorectal glandular architecture, assessed translational relevance and tested effects of 1,25(OH)2D3,theactive form of vitamin D. CM evolution was driven by oncogenic perturbation of the apical polarity (AP) complex comprising PTEN, CDC42 and PRKCZ (phosphatase and tensin homolog, cell division cycle 42 and protein kinase C zeta). Suppression of AP genes initiated a spatiotemporal cascade of mitotic spindle misorientation, apical membrane misalignment and aberrant epithelial configuration. Collectively, these events promoted "Swiss cheese-like" cribriform morphology (CM) comprising multiple abnormal "back to back" lumens surrounded by atypical stratified epithelium, in 3D colorectal gland models. Intestinal cancer driven purely by PTEN-deficiency in transgenic mice developed CM and in human CRC, CM associated with PTEN and PRKCZ readouts. Treatment of PTEN-deficient 3D cultures with 1,25(OH)2D3 upregulated PTEN, rapidly activated CDC42 and PRKCZ, corrected mitotic spindle alignment and suppressed CM development. Conversely, mutationally-activated KRAS blocked1,25(OH)2D3 rescue of glandular architecture. We conclude that 1,25(OH)2D3 upregulates AP signalling to reverse CM in a KRAS wild type (wt), clinically predictive CRC model system. Vitamin D could be developed as therapy to suppress inception or progression of a subset of colorectal tumors.
Collapse
Affiliation(s)
- Ravi K. Deevi
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Jane McClements
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Karen D. McCloskey
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Aliya Fatehullah
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Dorota Tkocz
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Arman Javadi
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Robyn Higginson
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | | | - Marnix Jansen
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK
| | - Alan Clarke
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, UK
| | - Maurice B. Loughrey
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queens University Belfast and Belfast Health and Social Care Trust, Belfast, UK
| | - Frederick C. Campbell
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| |
Collapse
|
22
|
Lili LN, Farkas AE, Gerner-Smidt C, Overgaard CE, Moreno CS, Parkos CA, Capaldo CT, Nusrat A. Claudin-based barrier differentiation in the colonic epithelial crypt niche involves Hopx/Klf4 and Tcf7l2/Hnf4-α cascades. Tissue Barriers 2016; 4:e1214038. [PMID: 27583195 DOI: 10.1080/21688370.2016.1214038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/11/2016] [Accepted: 07/11/2016] [Indexed: 12/13/2022] Open
Abstract
Colonic enterocytes form a rapidly renewing epithelium and barrier to luminal antigens. During renewal, coordinated expression of the claudin family of genes is vital to maintain the epithelial barrier. Disruption of this process contributes to barrier compromise and mucosal inflammatory diseases. However, little is known about the regulation of this critical aspect of epithelial cell differentiation. In order to identify claudin regulatory factors we utilized high-throughput gene microarrays and correlation analyses. We identified complex expression gradients for the transcription factors Hopx, Hnf4a, Klf4 and Tcf7l2, as well as 12 claudins, during differentiation. In vitro confirmatory methods identified 2 pathways that stimulate claudin expression; Hopx/Klf4 activation of Cldn4, 7 and 15, and Tcf7l2/Hnf4a up-regulation of Cldn23. Chromatin immunoprecipitation confirmed a Tcf7l2/Hnf4a/Claudin23 cascade. Furthermore, Hnf4a conditional knockout mice fail to induce Cldn23 during colonocyte differentiation. In conclusion, we report a comprehensive screen of colonic claudin gene expression and discover spatiotemporal Hopx/Klf4 and Tcf7l2/Hnf4a signaling as stimulators of colonic epithelial barrier differentiation.
Collapse
Affiliation(s)
- Loukia N Lili
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA, USA
| | - Attila E Farkas
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Institute of Biophysics, Biological Research Center, Szeged, Hungary
| | - Christian Gerner-Smidt
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA, USA
| | | | - Carlos S Moreno
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA, USA
| | - Charles A Parkos
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Asma Nusrat
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
23
|
He P, Zhao L, No YR, Karvar S, Yun CC. The NHERF1 PDZ1 domain and IRBIT interact and mediate the activation of Na+/H+ exchanger 3 by ANG II. Am J Physiol Renal Physiol 2016; 311:F343-51. [PMID: 27279487 DOI: 10.1152/ajprenal.00247.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/01/2016] [Indexed: 11/22/2022] Open
Abstract
Na(+)/H(+) exchanger (NHE)3, a major Na(+) transporter in the luminal membrane of the proximal tubule, is subject to ANG II regulation in renal Na(+)/fluid absorption and blood pressure control. We have previously shown that inositol 1,4,5-trisphosphate receptor-binding protein released with inositol 1,4,5-trisphosphate (IRBIT) mediates ANG II-induced exocytosis of NHE3 in cultured proximal tubule epithelial cells. In searching for scaffold protein(s) that coordinates with IRBIT in NHE3 trafficking, we found that NHE regulatory factor (NHERF)1, NHE3, and IRBIT proteins were coexpressed in the same macrocomplexes and that loss of ANG II type 1 receptors decreased their expression in the renal brush-border membrane. We found that NHERF1 was required for ANG II-mediated forward trafficking and activation of NHE3 in cultured cells. ANG II induced a concomitant increase of NHERF1 interactions with NHE3 and IRBIT, which were abolished when the NHERF1 PDZ1 domain was removed. Overexpression of a yellow fluorescent protein-NHERF1 construct that lacks PDZ1, but not PDZ2, failed to exaggerate the ANG II-dependent increase of NHE3 expression in the apical membrane. Moreover, exogenous expression of PDZ1 exerted a dominant negative effect on NHE3 activation by ANG II. We further demonstrated that IRBIT was indispensable for the ANG II-provoked increase in NHERF1-NHE3 interactions and that phosphorylation of IRBIT at Ser(68) was necessary for the assembly of the NHEF1-IRBIT-NHE3 complex. Taken together, our findings suggest that NHERF1 mediates ANG II-induced activation of renal NHE3, which requires coordination between IRBIT and the NHERF1 PDZ1 domain in binding and transporting NHE3.
Collapse
Affiliation(s)
- Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia;
| | - Luqing Zhao
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Gastroenterology, Beijing Hospital of Traditional Chinese Medicine affiliated with Capital Medical University, Beijing, China
| | - Yi Ran No
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Serhan Karvar
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Atlanta Veterans Affairs Medical Center, Decatur, Georgia; and Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
24
|
Foulke-Abel J, In J, Yin J, Zachos NC, Kovbasnjuk O, Estes MK, de Jonge H, Donowitz M. Human Enteroids as a Model of Upper Small Intestinal Ion Transport Physiology and Pathophysiology. Gastroenterology 2016; 150:638-649.e8. [PMID: 26677983 PMCID: PMC4766025 DOI: 10.1053/j.gastro.2015.11.047] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 11/05/2015] [Accepted: 11/25/2015] [Indexed: 01/18/2023]
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.
Collapse
Affiliation(s)
- Jennifer Foulke-Abel
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Julie In
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jianyi Yin
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicholas C Zachos
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Olga Kovbasnjuk
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Hugo de Jonge
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
25
|
Vogel GF, Klee KMC, Janecke AR, Müller T, Hess MW, Huber LA. Cargo-selective apical exocytosis in epithelial cells is conducted by Myo5B, Slp4a, Vamp7, and Syntaxin 3. J Cell Biol 2016; 211:587-604. [PMID: 26553929 PMCID: PMC4639860 DOI: 10.1083/jcb.201506112] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The motor protein Myo5B and t-SNARE Stx3 drive cargo-selective apical exocytosis in polarized epithelial cells in a pathway dependent on v-SNARE–like Slp4a, v-SNARE Vamp7, Sec1/Munc18-like protein Munc18-2, and the Rab11/8 cascade. Mutations in the motor protein Myosin Vb (Myo5B) or the soluble NSF attachment protein receptor Syntaxin 3 (Stx3) disturb epithelial polarity and cause microvillus inclusion disease (MVID), a lethal hereditary enteropathy affecting neonates. To understand the molecular mechanism of Myo5B and Stx3 interplay, we used genome editing to introduce a defined Myo5B patient mutation in a human epithelial cell line. Our results demonstrate a selective role of Myo5B and Stx3 for apical cargo exocytosis in polarized epithelial cells. Apical exocytosis of NHE3, CFTR (cystic fibrosis transmembrane conductance regulator), and GLUT5 required an interaction cascade of Rab11, Myo5B, Slp4a, Munc18-2, and Vamp7 with Stx3, which cooperate in the final steps of this selective apical traffic pathway. The brush border enzymes DPPIV and sucrase-isomaltase still correctly localize at the apical plasma membrane independent of this pathway. Hence, our work demonstrates how Myo5B, Stx3, Slp4a, Vamp7, Munc18-2, and Rab8/11 cooperate during selective apical cargo trafficking and exocytosis in epithelial cells and thereby provides further insight into MVID pathophysiology.
Collapse
Affiliation(s)
- Georg F Vogel
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria Division of Histology and Embryology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Katharina M C Klee
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria Institute of Molecular Biology, University of Innsbruck, 6020 Innsbruck, Austria Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
26
|
Chen L, Tuo B, Dong H. Regulation of Intestinal Glucose Absorption by Ion Channels and Transporters. Nutrients 2016; 8:nu8010043. [PMID: 26784222 PMCID: PMC4728656 DOI: 10.3390/nu8010043] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/18/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022] Open
Abstract
The absorption of glucose is electrogenic in the small intestinal epithelium. The major route for the transport of dietary glucose from intestinal lumen into enterocytes is the Na+/glucose cotransporter (SGLT1), although glucose transporter type 2 (GLUT2) may also play a role. The membrane potential of small intestinal epithelial cells (IEC) is important to regulate the activity of SGLT1. The maintenance of membrane potential mainly depends on the activities of cation channels and transporters. While the importance of SGLT1 in glucose absorption has been systemically studied in detail, little is currently known about the regulation of SGLT1 activity by cation channels and transporters. A growing line of evidence suggests that cytosolic calcium ([Ca2+]cyt) can regulate the absorption of glucose by adjusting GLUT2 and SGLT1. Moreover, the absorption of glucose and homeostasis of Ca2+ in IEC are regulated by cation channels and transporters, such as Ca2+ channels, K+ channels, Na+/Ca2+ exchangers, and Na+/H+ exchangers. In this review, we consider the involvement of these cation channels and transporters in the regulation of glucose uptake in the small intestine. Modulation of them may be a potential strategy for the management of obesity and diabetes.
Collapse
Affiliation(s)
- Lihong Chen
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China.
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China.
| | - Hui Dong
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China.
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
27
|
He P, Zhao L, Zhu L, Weinman EJ, De Giorgio R, Koval M, Srinivasan S, Yun CC. Restoration of Na+/H+ exchanger NHE3-containing macrocomplexes ameliorates diabetes-associated fluid loss. J Clin Invest 2015; 125:3519-31. [PMID: 26258413 DOI: 10.1172/jci79552] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 06/25/2015] [Indexed: 01/19/2023] Open
Abstract
Diarrhea is one of the troublesome complications of diabetes, and the underlying causes of this problem are complex. Here, we investigated whether altered electrolyte transport contributes to diabetic diarrhea. We found that the expression of Na+/H+ exchanger NHE3 and several scaffold proteins, including NHE3 regulatory factors (NHERFs), inositol trisphosphate (IP₃) receptor-binding protein released with IP₃ (IRBIT), and ezrin, was decreased in the intestinal brush border membrane (BBM) of mice with streptozotocin-induced diabetes. Treatment of diabetic mice with insulin restored intestinal NHE3 activity and fluid absorption. Molecular analysis revealed that NHE3, NHERF1, IRBIT, and ezrin form macrocomplexes, which are perturbed under diabetic conditions, and insulin administration reconstituted these macrocomplexes and restored NHE3 expression in the BBM. Silencing of NHERF1 or IRBIT prevented NHE3 trafficking to the BBM and insulin-dependent NHE3 activation. IRBIT facilitated the interaction of NHE3 with NHERF1 via protein kinase D2-dependent phosphorylation. Insulin stimulated ezrin phosphorylation, which enhanced the interaction of ezrin with NHERF1, IRBIT, and NHE3. Additionally, oral administration of lysophosphatidic acid (LPA) increased NHE3 activity and fluid absorption in diabetic mice via an insulin-independent pathway. Together, these findings indicate the importance of NHE3 in diabetic diarrhea and suggest LPA administration as a potential therapeutic strategy for management of diabetic diarrhea.
Collapse
|
28
|
Singh V, Yang J, Cha B, Chen TE, Sarker R, Yin J, Avula LR, Tse M, Donowitz M. Sorting nexin 27 regulates basal and stimulated brush border trafficking of NHE3. Mol Biol Cell 2015; 26:2030-43. [PMID: 25851603 PMCID: PMC4472014 DOI: 10.1091/mbc.e14-12-1597] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/01/2015] [Indexed: 12/21/2022] Open
Abstract
In polarized epithelial cells, SNX27 regulates PDZ domain–directed trafficking of NHE3 from endosomes to the plasma membrane and increases the stability of brush border NHE3. This establishes SNX27 as an important regulator of polarized sorting in epithelial cells. Sorting nexin 27 (SNX27) contains a PDZ domain that is phylogenetically related to the PDZ domains of the NHERF proteins. Studies on nonepithelial cells have shown that this protein is located in endosomes, where it regulates trafficking of cargo proteins in a PDZ domain–dependent manner. However, the role of SNX27 in trafficking of cargo proteins in epithelial cells has not been adequately explored. Here we show that SNX27 directly interacts with NHE3 (C-terminus) primarily through the SNX27 PDZ domain. A combination of knockdown and reconstitution experiments with wild type and a PDZ domain mutant (GYGF → GAGA) of SNX27 demonstrate that the PDZ domain of SNX27 is required to maintain basal NHE3 activity and surface expression of NHE3 in polarized epithelial cells. Biotinylation-based recycling and degradation studies in intestinal epithelial cells show that SNX27 is required for the exocytosis (not endocytosis) of NHE3 from early endosome to plasma membrane. SNX27 is also required to regulate the retention of NHE3 on the plasma membrane. The findings of the present study extend our understanding of PDZ-mediated recycling of cargo proteins from endosome to plasma membrane in epithelial cells.
Collapse
Affiliation(s)
- Varsha Singh
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jianbo Yang
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Boyoung Cha
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Tiane-e Chen
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Rafiquel Sarker
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jianyi Yin
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Leela Rani Avula
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ming Tse
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Mark Donowitz
- Gastroenterology Division, Departments of Physiology and Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
29
|
No YR, He P, Yoo BK, Yun CC. Unique regulation of human Na+/H+ exchanger 3 (NHE3) by Nedd4-2 ligase that differs from non-primate NHE3s. J Biol Chem 2014; 289:18360-72. [PMID: 24831004 DOI: 10.1074/jbc.m113.541706] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Na(+)/H(+) exchanger NHE3 expressed in the intestine and kidney plays a major role in NaCl and HCO3 (-) absorption that is closely linked to fluid absorption and blood pressure regulation. The Nedd4 family of E3 ubiquitin ligases interacts with a number of transporters and channels via PY motifs. A comparison of NHE3 sequences revealed the presence of PY motifs in NHE3s from human and several non-human primates but not in non-primate NHE3s. In this study we evaluated the differences between human and non-primate NHE3s in ubiquitination and interaction with Nedd4-2. We found that Nedd4-2 ubiquitinated human NHE3 (hNHE3) and altered its expression and activity. Surprisingly, rat NHE3 co-immunoprecipitated Nedd4-2, but its expression and activity were not altered by silencing of Nedd4-2. Ubiquitination by Nedd4-2 rendered hNHE3 to undergo internalization at a significantly greater rate than non-primate NHE3s without altering protein stability. Insertion of a PY motif in rabbit NHE3 recapitulated the interaction with Nedd4-2 and enhanced internalization. Thus, we propose a new model where disruption of Nedd4-2 interaction elevates hNHE3 expression and activity.
Collapse
Affiliation(s)
- Yi Ran No
- From the Division of Digestive Diseases, Department of Medicine and
| | - Peijian He
- From the Division of Digestive Diseases, Department of Medicine and
| | - Byong Kwon Yoo
- From the Division of Digestive Diseases, Department of Medicine and
| | - C Chris Yun
- From the Division of Digestive Diseases, Department of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
30
|
Muthusamy S, Cheng M, Jeong JJ, Kumar A, Dudeja PK, Malakooti J. Extracellular acidosis stimulates NHE2 expression through activation of transcription factor Egr-1 in the intestinal epithelial cells. PLoS One 2013; 8:e82023. [PMID: 24376510 PMCID: PMC3871166 DOI: 10.1371/journal.pone.0082023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/29/2013] [Indexed: 01/28/2023] Open
Abstract
Na(+)/H(+) exchangers (NHEs) play important roles in regulating internal pH (pHi), cell volume and neutral Na(+) absorption in the human intestine. Earlier studies have shown that low extracellular pH (pHe) and metabolic acidosis increases the expression and function of NHE1-3 genes. However, transcriptional mechanisms involved remained unknown. Therefore, we investigated the molecular mechanisms underlying acid-induced NHE2 expression in C2BBe1 and SK-CO15 intestinal epithelial cells. Assessing total RNA and protein by RT-PCR and Western blot analysis, respectively, displayed significant increases in the NHE2 mRNA and protein levels in cells exposed to acidic media (pH 6.5 and 6.7) compared to normal medium. Acid treatment was also associated with a significant enhancement in NHE2 transport activity. Quantification of the heterogeneous nuclear RNA indicated that the rate of NHE2 transcription was increased in response to acid. Furthermore, acid caused a significant increase in NHE2 promoter activity confirming transcriptional upregulation. Through functional and mutational studies the acid-response element was mapped to a 15-nucleotide GC-rich sequence at bp -337 to -323 upstream from the transcription start site. We previously identified this element as an overlapping Egr-1/Sp1/Egr-1 motif that was essential for the NHE2 upregulation by mitogen-induced transcription factor Egr-1. Cells exposed to acid exhibited a temporal increase in Egr-1 mRNA and protein expression. These events were followed by Egr-1 nuclear accumulation, as detected by immunofluorescence microscopy, and potentiated its in vitro and in vivo interaction with the NHE2 promoter. Disruption of ESE motif and knockdown of Egr-1 expression by targeted small interfering RNA abrogated the acid-induced NHE2 transcriptional activity. These data indicate that the acid-dependent NHE2 stimulation is implemented by transcriptional upregulation of NHE2 via acid-induced Egr-1 in the intestinal epithelial cells.
Collapse
Affiliation(s)
- Saminathan Muthusamy
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ming Cheng
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jong-Jin Jeong
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Pradeep K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Jaleh Malakooti
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
31
|
Singh V, Raheja G, Borthakur A, Kumar A, Gill RK, Alakkam A, Malakooti J, Dudeja PK. Lactobacillus acidophilus upregulates intestinal NHE3 expression and function. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1393-401. [PMID: 23086913 PMCID: PMC3532544 DOI: 10.1152/ajpgi.00345.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A major mechanism of electroneutral NaCl absorption in the human ileum and colon involves coupling of Na(+)/H(+) and Cl(-)/HCO(3)(-) exchangers. Disturbances in these mechanisms have been implicated in diarrheal conditions. Probiotics such as Lactobacillus have been indicated to be beneficial in the management of gastrointestinal disorders, including diarrhea. However, the molecular mechanisms underlying antidiarrheal effects of probiotics have not been fully understood. We have previously demonstrated Lactobacillus acidophilus (LA) to stimulate Cl(-)/HCO3- exchange activity via an increase in the surface levels and expression of the Cl(-)/HCO3- exchanger DRA in vitro and in vivo. However, the effects of LA on NHE3, the Na(+)/H(+) exchanger involved in the coupled electroneutral NaCl absorption, are not known. Current studies were, therefore, undertaken to investigate the effects of LA on the function and expression of NHE3 and to determine the mechanisms involved. Treatment of Caco2 cells with LA or its conditioned culture supernatant (CS) for 8-24 h resulted in a significant increase in Na(+)/H(+) exchange activity, mRNA, and protein levels of NHE3. LA-CS upregulation of NHE3 function and expression was also observed in SK-CO15 cells, a human colonic adenocarcinoma cell line. Additionally, LA treatment increased NHE3 promoter activity, suggesting involvement of transcriptional mechanisms. In vivo, mice gavaged with live LA showed significant increase in NHE3 mRNA and protein expression in the ileum and colonic regions. In conclusion, LA-induced increase in NHE3 expression may contribute to the upregulation of intestinal electrolyte absorption and might underlie the potential antidiarrheal effects of probiotics.
Collapse
Affiliation(s)
- Varsha Singh
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Geetu Raheja
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Alip Borthakur
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Anoop Kumar
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Ravinder K. Gill
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Anas Alakkam
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Jaleh Malakooti
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| | - Pradeep K. Dudeja
- Section of Digestive Diseases and Nutrition, Department of Medicine, University of Illinois at Chicago and Jesse Brown VA Medical Center, Chicago, Illinois
| |
Collapse
|