1
|
Dooley SA, Kolobova E, Burman A, Kaji I, Digrazia JR, Stubler R, Goldstein A, Packirisamy C, Coutts AW, Saqui-Salces M, Gao N, Engevik MA, Shub MD, Goldenring JR, Engevik AC. Myosin Vb Traffics P-Glycoprotein to the Apical Membrane of Intestinal Epithelial Cells. Gastroenterology 2025; 168:84-98.e9. [PMID: 39299401 DOI: 10.1053/j.gastro.2024.09.007] [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: 05/10/2022] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND & AIMS The xenobiotic efflux pump P-glycoprotein is highly expressed on the apical membrane of the gastrointestinal tract, where it regulates the levels of intracellular substrates. P-glycoprotein is altered in disease, but the mechanisms that regulate the levels of P-glycoprotein are still being explored. The molecular motor myosin Vb (Myo5b) traffics diverse cargo to the apical membrane of intestinal epithelial cells. We hypothesized that Myo5b was responsible for the delivery of P-glycoprotein to the apical membrane of enterocytes. METHODS We used multiple murine models that lack functional Myo5b or the myosin binding partner Rab11a to analyze P-glycoprotein localization. Pig and human tissue were analyzed to determine P-glycoprotein localization in the setting of MYO5B mutations. Intestinal organoids were used to examine P-glycoprotein trafficking and to assay P-glycoprotein function when MYO5 is inhibited. RESULTS In mice lacking Myo5b or the binding partner Rab11a, P-glycoprotein was improperly trafficked and had decreased presence in the brush border of enterocytes. Immunostaining of a pig model lacking functional Myo5b and human biopsies from a patient with an inactivating mutation in Myo5b also showed altered localization of intestinal P-glycoprotein. Human intestinal organoids expressing the motorless MYO5B tail domain had colocalization with P-glycoprotein, confirming that P-glycoprotein was trafficked by MYO5B in human enterocytes. Inhibition of MYO5 in human intestinal cell lines and organoids resulted in decreased P-glycoprotein capacity. Additionally, inhibition of MYO5 in human colon cancer cells diminished P-glycoprotein activity and increased cell death in response to a chemotherapeutic drug. CONCLUSIONS Collectively, these data demonstrate that Myo5b is necessary for the apical delivery of P-glycoprotein.
Collapse
Affiliation(s)
- Sarah A Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Elena Kolobova
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Andreanna Burman
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jessica R Digrazia
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Rachel Stubler
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Anna Goldstein
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charulekha Packirisamy
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | | | - Milena Saqui-Salces
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota
| | - Nan Gao
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Melinda A Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Mitchell D Shub
- Division of Gastroenterology, Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | - Amy C Engevik
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
2
|
Kaji I, Thiagarajah JR, Goldenring JR. Modeling the cell biology of monogenetic intestinal epithelial disorders. J Cell Biol 2024; 223:e202310118. [PMID: 38683247 PMCID: PMC11058565 DOI: 10.1083/jcb.202310118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Monogenetic variants are responsible for a range of congenital human diseases. Variants in genes that are important for intestinal epithelial function cause a group of disorders characterized by severe diarrhea and loss of nutrient absorption called congenital diarrheas and enteropathies (CODEs). CODE-causing genes include nutrient transporters, enzymes, structural proteins, and vesicular trafficking proteins in intestinal epithelial cells. Several severe CODE disorders result from the loss-of-function in key regulators of polarized endocytic trafficking such as the motor protein, Myosin VB (MYO5B), as well as STX3, STXBP2, and UNC45A. Investigations of the cell biology and pathophysiology following loss-of-function in these genes have led to an increased understanding of both homeostatic and pathological vesicular trafficking in intestinal epithelial cells. Modeling different CODEs through investigation of changes in patient tissues, coupled with the development of animal models and patient-derived enteroids, has provided critical insights into the enterocyte differentiation and function. Linking basic knowledge of cell biology with the phenotype of specific patient variants is a key step in developing effective treatments for rare monogenetic diseases. This knowledge can also be applied more broadly to our understanding of common epithelial disorders.
Collapse
Affiliation(s)
- Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jay R. Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, MA, USA
- Harvard Digestive Disease Center, Boston, MA, USA
| | - James R. Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Nashville VA Medical Center, Nashville, TN, USA
| |
Collapse
|
3
|
Carlos dos Reis D, Dastoor P, Santos AK, Sumigray K, Ameen NA. CFTR high expresser cells in cystic fibrosis and intestinal diseases. Heliyon 2023; 9:e14568. [PMID: 36967909 PMCID: PMC10031467 DOI: 10.1016/j.heliyon.2023.e14568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), the Cl-/HCO3 - channel implicated in Cystic Fibrosis, is critical to the pathophysiology of many gastrointestinal diseases. Defects in CFTR lead to intestinal dysfunction, malabsorption, obstruction, infection, inflammation, and cancer that increases morbidity and reduces quality of life. This review will focus on CFTR in the intestine and the implications of the subpopulation of CFTR High Expresser Cells (CHEs) in Cystic Fibrosis (CF), intestinal physiology and pathophysiology of intestinal diseases.
Collapse
Affiliation(s)
- Diego Carlos dos Reis
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
| | - Parinaz Dastoor
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
| | - Anderson Kenedy Santos
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Kaelyn Sumigray
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06510, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Nadia A. Ameen
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT06510, USA
- Corresponding author. Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA.
| |
Collapse
|
4
|
Burman A, Momoh M, Sampson L, Skelton J, Roland JT, Ramos C, Krystofiak E, Acra S, Goldenring JR, Kaji I. Modeling of a Novel Patient-Based MYO5B Point Mutation Reveals Insights Into MVID Pathogenesis. Cell Mol Gastroenterol Hepatol 2022; 15:1022-1026. [PMID: 36592862 PMCID: PMC10041088 DOI: 10.1016/j.jcmgh.2022.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Affiliation(s)
- Andreanna Burman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Momoh
- Epithelial Biology Center, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leesa Sampson
- Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee
| | - Jennifer Skelton
- Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee
| | - Joseph T Roland
- Epithelial Biology Center, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cynthia Ramos
- Epithelial Biology Center, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Evan Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Sari Acra
- Division of Pediatric Gastroenterology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James R Goldenring
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Epithelial Biology Center, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee
| | - Izumi Kaji
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Epithelial Biology Center, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
5
|
Cartón-García F, Brotons B, Anguita E, Dopeso H, Tarragona J, Nieto R, García-Vidal E, Macaya I, Zagyva Z, Dalmau M, Sánchez-Martín M, van Ijzendoorn SCD, Landolfi S, Hernandez-Losa J, Schwartz Jr S, Matias-Guiu X, Ramón y Cajal S, Martínez-Barriocanal Á, Arango D. Myosin Vb as a tumor suppressor gene in intestinal cancer. Oncogene 2022; 41:5279-5288. [DOI: 10.1038/s41388-022-02508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
|
6
|
Sun Y, Leng C, van Ijzendoorn SCD. Fetal Bowel Abnormalities Suspected by Ultrasonography in Microvillus Inclusion Disease: Prevalence and Clinical Significance. J Clin Med 2022; 11:jcm11154331. [PMID: 35893420 PMCID: PMC9332086 DOI: 10.3390/jcm11154331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/28/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023] Open
Abstract
Microvillus inclusion disease (MVID) is a rare, inherited, congenital, diarrheal disorder that is invariably fatal if left untreated. Within days after birth, MVID presents as a life-threatening emergency characterized by severe dehydration, metabolic acidosis, and weight loss. Diagnosis is cumbersome and can take a long time. Whether MVID could be diagnosed before birth is not known. Anecdotal reports of MVID-associated fetal bowel abnormalities suspected by ultrasonography (that is, dilated bowel loops and polyhydramnios) have been published. These are believed to be rare, but their prevalence in MVID has not been investigated. Here, we have performed a comprehensive retrospective study of 117 published MVID cases spanning three decades. We find that fetal bowel abnormalities in MVID occurred in up to 60% of cases of MVID for which prenatal ultrasonography or pregnancy details were reported. Suspected fetal bowel abnormalities appeared in the third trimester of pregnancy and correlated with postnatal, early-onset diarrhea and case-fatality risk during infancy. Fetal bowel dilation correlated with MYO5B loss-of-function variants. In conclusion, MVID has already started during fetal life in a significant number of cases. Genetic testing for MVID-causing gene variants in cases where fetal bowel abnormalities are suspected by ultrasonography may allow for the prenatal diagnosis of MVID in a significant percentage of cases, enabling optimal preparation for neonatal intensive care.
Collapse
Affiliation(s)
- Yue Sun
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (Y.S.); (C.L.)
- Center for Liver, Digestive & Metabolic Disease, University of Groningen, University Medical Center Groningen, 9700 AD Groningen, The Netherlands
| | - Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (Y.S.); (C.L.)
- Center for Liver, Digestive & Metabolic Disease, University of Groningen, University Medical Center Groningen, 9700 AD Groningen, The Netherlands
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Department of Thoracic Surgery, Sun Yat-sen University Cancer Centre, Guangzhou 510060, China
| | - Sven C. D. van Ijzendoorn
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands; (Y.S.); (C.L.)
- Center for Liver, Digestive & Metabolic Disease, University of Groningen, University Medical Center Groningen, 9700 AD Groningen, The Netherlands
- Correspondence:
| |
Collapse
|
7
|
Engevik MA, Engevik AC. Myosins and membrane trafficking in intestinal brush border assembly. Curr Opin Cell Biol 2022; 77:102117. [PMID: 35870341 DOI: 10.1016/j.ceb.2022.102117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
Myosins are a class of motors that participate in a wide variety of cellular functions including organelle transport, cell adhesion, endocytosis and exocytosis, movement of RNA, and cell motility. Among the emerging roles for myosins is regulation of the assembly, morphology, and function of actin protrusions such as microvilli. The intestine harbors an elaborate apical membrane composed of highly organized microvilli. Microvilli assembly and function are intricately tied to several myosins including Myosin 1a, non-muscle Myosin 2c, Myosin 5b, Myosin 6, and Myosin 7b. Here, we review the research progress made in our understanding of myosin mediated apical assembly.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina
| | - Amy C Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina.
| |
Collapse
|
8
|
Ahsan MK, dos Reis DC, Barbieri A, Sumigray KD, Nottoli T, Salas PJ, Ameen NA. Loss of Serum Glucocorticoid-Inducible Kinase 1 SGK1 Worsens Malabsorption and Diarrhea in Microvillus Inclusion Disease (MVID). J Clin Med 2022; 11:jcm11144179. [PMID: 35887942 PMCID: PMC9319011 DOI: 10.3390/jcm11144179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Microvillus inclusion disease (MVID), a lethal congenital diarrheal disease, results from loss of function mutations in the apical actin motor myosin VB (MYO5B). How loss of MYO5B leads to both malabsorption and fluid secretion is not well understood. Serum glucocorticoid-inducible kinase 1 (SGK1) regulates intestinal carbohydrate and ion transporters including cystic fibrosis transmembrane conductance regulator (CFTR). We hypothesized that loss of SGK1 could reduce CFTR fluid secretion and MVID diarrhea. Using CRISPR-Cas9 approaches, we generated R26CreER;MYO5Bf/f conditional single knockout (cMYO5BKO) and R26CreER;MYO5Bf/f;SGK1f/f double knockout (cSGK1/MYO5B-DKO) mice. Tamoxifen-treated cMYO5BKO mice resulted in characteristic features of human MVID including severe diarrhea, microvillus inclusions (MIs) in enterocytes, defective apical traffic, and depolarization of transporters. However, apical CFTR distribution was preserved in crypts and depolarized in villus enterocytes, and CFTR high expresser (CHE) cells were observed. cMYO5BKO mice displayed increased phosphorylation of SGK1, PDK1, and the PDK1 target PKCι in the intestine. Surprisingly, tamoxifen-treated cSGK1/MYO5B-DKO mice displayed more severe diarrhea than cMYO5BKO, with preservation of apical CFTR and CHE cells, greater fecal glucose and reduced SGLT1 and GLUT2 in the intestine. We conclude that loss of SGK1 worsens carbohydrate malabsorption and diarrhea in MVID.
Collapse
Affiliation(s)
- Md Kaimul Ahsan
- Department of Pediatrics, Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT 06510, USA; (M.K.A.); (D.C.d.R.)
| | - Diego Carlos dos Reis
- Department of Pediatrics, Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT 06510, USA; (M.K.A.); (D.C.d.R.)
| | - Andrea Barbieri
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Kaelyn D. Sumigray
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Timothy Nottoli
- Genome Editing Center, Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Pedro J. Salas
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, FL 33146, USA;
| | - Nadia A. Ameen
- Department of Pediatrics, Gastroenterology and Hepatology, Yale University School of Medicine, New Haven, CT 06510, USA; (M.K.A.); (D.C.d.R.)
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Correspondence:
| |
Collapse
|
9
|
Bowman DM, Kaji I, Goldenring JR. Altered MYO5B Function Underlies Microvillus Inclusion Disease: Opportunities for Intervention at a Cellular Level. Cell Mol Gastroenterol Hepatol 2022; 14:553-565. [PMID: 35660026 PMCID: PMC9304615 DOI: 10.1016/j.jcmgh.2022.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/31/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022]
Abstract
Microvillus inclusion disease (MVID) is a congenital diarrheal disorder resulting in life-threatening secretory diarrhea in newborns. Inactivating and nonsense mutations in myosin Vb (MYO5B) have been identified in MVID patients. Work using patient tissues, cell lines, mice, and pigs has led to critical insights into the pathology of MVID and a better understanding of both apical trafficking in intestinal enterocytes and intestinal stem cell differentiation. These studies have demonstrated that loss of MYO5B or inactivating mutations lead to loss of apical sodium and water transporters, without loss of apical CFTR, accounting for the major pathology of the disease. In addition, loss of MYO5B expression induces the formation of microvillus inclusions through apical bulk endocytosis that utilizes dynamin and PACSIN2 and recruits tight junction proteins to the sites of bulk endosome formation. Importantly, formation of microvillus inclusions is not required for the induction of diarrhea. Recent investigations have demonstrated that administration of lysophosphatidic acid (LPA) can partially reestablish apical ion transporters in enterocytes of MYO5B KO mice. In addition, further studies have shown that MYO5B loss induces an imbalance in Wnt/Notch signaling pathways that can lead to alterations in enterocyte maturation and tuft cell lineage differentiation. Inhibition of Notch signaling leads to improvements in those cell differentiation deficits. These studies demonstrate that directed strategies through LPA receptor activation and Notch inhibition can bypass the inhibitory effects of MYO5B loss. Thus, effective strategies may be successful in MVID patients and other congenital diarrhea syndromes to reestablish proper apical membrane absorption of sodium and water in enterocytes and ameliorate life-threatening congenital diarrhea.
Collapse
Affiliation(s)
- Deanna M Bowman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee.
| |
Collapse
|
10
|
Li Q, Zhou Z, Sun Y, Sun C, Klappe K, van IJzendoorn SC. A Functional Relationship Between UNC45A and MYO5B Connects Two Rare Diseases With Shared Enteropathy. Cell Mol Gastroenterol Hepatol 2022; 14:295-310. [PMID: 35421597 PMCID: PMC9218578 DOI: 10.1016/j.jcmgh.2022.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND & AIMS UNC45A is a myosin (co-)chaperone, and mutations in the UNC45A gene were recently identified in osteo-oto-hepato-enteric (O2HE) syndrome patients presenting with congenital diarrhea and intrahepatic cholestasis. Congenital diarrhea and intrahepatic cholestasis are also the prime symptoms in patients with microvillus inclusion disease (MVID) and mutations in MYO5B, encoding the recycling endosome-associated myosin Vb. The aim of this study was to determine whether UNC45A and myosin Vb are functionally linked. METHODS CRISPR-Cas9 gene editing and site-directed mutagenesis were performed with intestinal epithelial and hepatocellular cell lines, followed by Western blotting, quantitative polymerase chain reaction, and scanning electron and/or confocal fluorescence microscopy to determine the relationship between (mutants of) UNC45A and myosin Vb. RESULTS UNC45A depletion in intestinal and hepatic cells reduced myosin Vb protein expression, and in intestinal epithelial cells, it affected 2 myosin Vb-dependent processes that underlie MVID pathogenesis: rat sarcoma-associated binding protein (RAB)11A-positve recycling endosome positioning and microvilli development. Reintroduction of UNC45A in UNC45A-depleted cells restored myosin Vb expression, and reintroduction of UNC45A or myosin Vb, but not the O2HE patient UNC45A-c.1268T>A variant, restored recycling endosome positioning and microvilli development. The O2HE patient-associated p.V423D substitution, encoded by the UNC45A-c.1268T>A variant, impaired UNC45A protein stability but as such not the ability of UNC45A to promote myosin Vb expression and microvilli development. CONCLUSIONS A functional relationship exists between UNC45A and myosin Vb, thereby connecting 2 rare congenital diseases with overlapping enteropathy at the molecular level. Protein instability rather than functional impairment underlies the pathogenicity of the O2HE syndrome-associated UNC45A-p.V423D mutation.
Collapse
Affiliation(s)
| | | | | | | | | | - Sven C.D. van IJzendoorn
- Correspondence Address correspondence to: Sven C. D. van IJzendoorn, PhD, Department of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
11
|
Leng C, Sun Y, van IJzendoorn SCD. Risk and Clinical Significance of Idiopathic Preterm Birth in Microvillus Inclusion Disease. J Clin Med 2021; 10:jcm10173935. [PMID: 34501384 PMCID: PMC8432107 DOI: 10.3390/jcm10173935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a rare enteropathy caused by mutations in the MYO5B or STX3 gene. MVID is a disease that is difficult to manage with clinical heterogeneity. Therefore, knowledge about factors influencing MVID morbidity and mortality is urgently needed. Triggered by a recent study that reported a high percentage of preterm births in twelve cases of MVID, we have conducted a comprehensive retrospective study involving 88 cases of MVID with reported gestational ages. We found that moderate to late preterm birth occurred in more than half of all cases, and this was particularly prominent in MYO5B-associated MVID. Preterm birth in MVID counterintuitively correlated with higher birth weight percentiles, and correlated with higher stool outputs and a significantly shorter average survival time. Data from this study thus demonstrate an increased risk of preterm birth in MYO5B-associated MVID, with a clinical impact on morbidity and mortality. Adverse effects associated with preterm birth should be taken into account in the care of children diagnosed with MVID. Documentation of gestational age may contribute to a better prognostic risk assessment in MVID.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, Centre for Liver, Digestive and Metabolic Disease, University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (C.L.); (Y.S.)
- Department of Thoracic Surgery, Guangdong Esophageal Cancer Institute, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Centre, Guangzhou 510060, China
| | - Yue Sun
- Department of Biomedical Sciences of Cells and Systems, Centre for Liver, Digestive and Metabolic Disease, University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (C.L.); (Y.S.)
| | - Sven C. D. van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, Centre for Liver, Digestive and Metabolic Disease, University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (C.L.); (Y.S.)
- Correspondence:
| |
Collapse
|
12
|
Das B, Okamoto K, Rabalais J, Young JA, Barrett KE, Sivagnanam M. Aberrant Epithelial Differentiation Contributes to Pathogenesis in a Murine Model of Congenital Tufting Enteropathy. Cell Mol Gastroenterol Hepatol 2021; 12:1353-1371. [PMID: 34198013 PMCID: PMC8479479 DOI: 10.1016/j.jcmgh.2021.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Congenital tufting enteropathy (CTE) is an intractable diarrheal disease of infancy caused by mutations of epithelial cell adhesion molecule (EpCAM). The cellular and molecular basis of CTE pathology has been elusive. We hypothesized that the loss of EpCAM in CTE results in altered lineage differentiation and defects in absorptive enterocytes thereby contributing to CTE pathogenesis. METHODS Intestine and colon from mice expressing a CTE-associated mutant form of EpCAM (mutant mice) were evaluated for specific markers by quantitative real-time polymerase chain reaction, Western blotting, and immunostaining. Body weight, blood glucose, and intestinal enzyme activity were also investigated. Enteroids derived from mutant mice were used to assess whether the decreased census of major secretory cells could be rescued. RESULTS Mutant mice exhibited alterations in brush-border ultrastructure, function, disaccharidase activity, and glucose absorption, potentially contributing to nutrient malabsorption and impaired weight gain. Altered cell differentiation in mutant mice led to decreased enteroendocrine cells and increased numbers of nonsecretory cells, though the hypertrophied absorptive enterocytes lacked key features, causing brush border malfunction. Further, treatment with the Notch signaling inhibitor, DAPT, increased the numbers of major secretory cell types in mutant enteroids (graphical abstract 1). CONCLUSIONS Alterations in intestinal epithelial cell differentiation in mutant mice favor an increase in absorptive cells at the expense of major secretory cells. Although the proportion of absorptive enterocytes is increased, they lack key functional properties. We conclude that these effects underlie pathogenic features of CTE such as malabsorption and diarrhea, and ultimately the failure to thrive seen in patients.
Collapse
Affiliation(s)
- Barun Das
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kevin Okamoto
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Rabalais
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Jocelyn A. Young
- Department of Pediatrics, University of California, San Diego, La Jolla, California,Department of Pediatrics, Rady Children’s Hospital, San Diego, California
| | - Kim E. Barrett
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Mamata Sivagnanam
- Department of Pediatrics, University of California, San Diego, La Jolla, California,Department of Pediatrics, Rady Children’s Hospital, San Diego, California,Correspondence Address correspondence to: Mamata Sivagnanam, MD, Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, 9500 Gilman Drive, La Jolla, CA 92093. fax: 858-967-8917.
| |
Collapse
|
13
|
Recruitment of Polarity Complexes and Tight Junction Proteins to the Site of Apical Bulk Endocytosis. Cell Mol Gastroenterol Hepatol 2021; 12:59-80. [PMID: 33548596 PMCID: PMC8082271 DOI: 10.1016/j.jcmgh.2021.01.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS The molecular motor, Myosin Vb (MYO5B), is well documented for its role in trafficking cargo to the apical membrane of epithelial cells. Despite its involvement in regulating apical proteins, the role of MYO5B in cell polarity is less clear. Inactivating mutations in MYO5B result in microvillus inclusion disease (MVID), a disorder characterized by loss of key apical transporters and the presence of intracellular inclusions in enterocytes. We previously identified that inclusions in Myo5b knockout (KO) mice form from invagination of the apical brush border via apical bulk endocytosis. Herein, we sought to elucidate the role of polarity complexes and tight junction proteins during the formation of inclusions. METHODS Intestinal tissue from neonatal control and Myo5b KO littermates was analyzed by immunofluorescence to determine the localization of polarity complexes and tight junction proteins. RESULTS Proteins that make up the apical polarity complexes-Crumbs3 and Pars complexes-were associated with inclusions in Myo5b KO mice. In addition, tight junction proteins were observed to be concentrated over inclusions that were present at the apical membrane of Myo5b-deficient enterocytes in vivo and in vitro. Our mouse findings are complemented by immunostaining in a large animal swine model of MVID genetically engineered to express a human MVID-associated mutation that shows an accumulation of Claudin-2 over forming inclusions. The findings from our swine model of MVID suggest that a similar mechanism of tight junction accumulation occurs in patients with MVID. CONCLUSIONS These data show that apical bulk endocytosis involves the altered localization of apical polarity proteins and tight junction proteins after loss of Myo5b.
Collapse
|
14
|
Leng C, Rings EHHM, de Wildt SN, van IJzendoorn SCD. Pharmacological and Parenteral Nutrition-Based Interventions in Microvillus Inclusion Disease. J Clin Med 2020; 10:jcm10010022. [PMID: 33374831 PMCID: PMC7794843 DOI: 10.3390/jcm10010022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a rare inherited and invariably fatal enteropathy, characterized by severe intractable secretory diarrhea and nutrient malabsorption. No cure exists, and patients typically die during infancy because of treatment-related complications. The need for alternative treatment strategies is evident. Several pharmacological interventions with variable successes have been tried and reported for individual patients as part of their clinical care. Unfortunately, these interventions and their outcomes have remained hidden in case reports and have not been reviewed. Further, recent advances regarding MVID pathogenesis have shed new light on the outcomes of these pharmacological interventions and offer suggestions for future clinical research and trials. Hence, an inventory of reported pharmacological interventions in MVID, their rationales and outcomes, and a discussion of these in the light of current knowledge is opportune. Together with a discussion on MVID-specific pharmacokinetic, -dynamic, and -genetic concerns that pose unique challenges regarding pharmacological strategies, we envision that this paper will aid researchers and clinicians in their efforts to develop pharmacological interventions to combat this devastating disease.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Edmond H. H. M. Rings
- Department of Pediatrics, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Saskia N. de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Intensive Care and Department of Pediatric Surgery, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Sven C. D. van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- Correspondence: ; Tel.: +31-(0)50-3616209
| |
Collapse
|
15
|
Ahsan MK, Figueroa-Hall L, Baratta V, Garcia-Milian R, Lam TT, Hoque K, Salas PJ, Ameen NA. Glucocorticoids and serum- and glucocorticoid-inducible kinase 1 are potent regulators of CFTR in the native intestine: implications for stress-induced diarrhea. Am J Physiol Gastrointest Liver Physiol 2020; 319:G121-G132. [PMID: 32567324 PMCID: PMC7500270 DOI: 10.1152/ajpgi.00076.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nongenomic glucocorticoid (GC) and serum- and glucocorticoid-inducible kinase 1 (SGK1) signaling regulate ion transport, but CFTR has not been investigated in the intestine. We examined GC, SGK1, and phosphatidylinositol 3-kinase (PI3K) kinase signaling of CFTR ion transport in native intestine and the role of GCs on mRNA, protein, surface expression, and cyclic guanosine monophosphate (cGMP)-elicited diarrhea. Rats were treated with dexamethasone (DEXA; 2 mg/kg ip) or DMSO for 1, 4, and 24 h. Cyclic adenosine monophosphate (cAMP)-activated ion transport was examined in the presence or absence of SGK1 and PI3K inhibitors. Phosphorylation of SGK1, phosphoinositide-dependent kinase 1, and Akt kinases was confirmed by immunoblots using phosphor-specific antibodies. Tissue lysates were analyzed by mass spectrometry. CFTR and SGK1 mRNA were measured by quantitative PCR. Changes in total and surface CFTR protein were determined. The role of GC in cGMP-activated CFTR ion transport was examined. GC synergistically increased CFTR ion transport by SGK1 and PI3K signaling and increased CFTR protein without altering SGK1 or CFTR mRNA. GC induced highest levels of CFTR protein at 4 h that were associated with marked increase in surface CFTR, phosphorylation of the ubiquitin ligase neural precursor cell expressed developmentally downregulated 4-like (Nedd4-2), and 14-3-3ε, supporting their roles in surface retention and stability. Coimmunoprecipitation of CFTR, Nedd4-2, and 14-3-3ε indicated that assembly of this complex is a likely effector of the SGK and Akt pathways. Mass spectrometry identified phosphorylated peptides in relevant proteins. GC-SGK1 potently regulates CFTR in the intestine and is implicated in diarrheal disease.NEW & NOTEWORTHY This is the first study to examine the mechanisms of glucocorticoid, serum- and glucocorticoid-inducible kinase 1, and nongenomic kinase signaling of CFTR in the native intestine. We identified unique and druggable intestine-specific factors of the pathway that are targets for treating stress-induced diarrhea.
Collapse
Affiliation(s)
- Md Kaimul Ahsan
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut
| | - Leandra Figueroa-Hall
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut
| | - Vanessa Baratta
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, Connecticut
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut.,Mass Spectrometry and Proteomics Resource, W. M. Keck Biotechnology Resource Laboratory, Yale University, New Haven, Connecticut
| | - Kazi Hoque
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Pedro J Salas
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Nadia A Ameen
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut.,Department of Pediatrics, Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
16
|
Engevik AC, Coutts AW, Kaji I, Rodriguez P, Ongaratto F, Saqui-Salces M, Medida RL, Meyer AR, Kolobova E, Engevik MA, Williams JA, Shub MD, Carlson DF, Melkamu T, Goldenring JR. Editing Myosin VB Gene to Create Porcine Model of Microvillus Inclusion Disease, With Microvillus-Lined Inclusions and Alterations in Sodium Transporters. Gastroenterology 2020; 158:2236-2249.e9. [PMID: 32112796 PMCID: PMC7282982 DOI: 10.1053/j.gastro.2020.02.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Microvillus inclusion disease (MVID) is caused by inactivating mutations in the myosin VB gene (MYO5B). MVID is a complex disorder characterized by chronic, watery, life-threatening diarrhea that usually begins in the first hours to days of life. We developed a large animal model of MVID to better understand its pathophysiology. METHODS Pigs were cloned by transfer of chromatin from swine primary fetal fibroblasts, which were edited with TALENs and single-strand oligonucleotide to introduce a P663-L663 substitution in the endogenous swine MYO5B (corresponding to the P660L mutation in human MYO5B, associated with MVID) to fertilized oocytes. We analyzed duodenal tissues from patients with MVID (with the MYO5B P660L mutation) and without (controls), and from pigs using immunohistochemistry. Enteroids were generated from pigs with MYO5B(P663L) and without the substitution (control pigs). RESULTS Duodenal tissues from patients with MVID lacked MYO5B at the base of the apical membrane of intestinal cells; instead MYO5B was intracellular. Intestinal tissues and derived enteroids from MYO5B(P663L) piglets had reduced apical levels and diffuse subapical levels of sodium hydrogen exchanger 3 and SGLT1, which regulate transport of sodium, glucose, and water, compared with tissues from control piglets. However, intestinal tissues and derived enteroids from MYO5B(P663L) piglets maintained CFTR on apical membranes, like tissues from control pigs. Liver tissues from MYO5B(P663L) piglets had alterations in bile salt export pump, a transporter that facilitates bile flow, which is normally expressed in the bile canaliculi in the liver. CONCLUSIONS We developed a large animal model of MVID that has many features of the human disease. Studies of this model could provide information about the functions of MYO5B and MVID pathogenesis, and might lead to new treatments.
Collapse
Affiliation(s)
- Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | | | - Izumi Kaji
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | - Milena Saqui-Salces
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota
| | - Ramya Lekha Medida
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota
| | - Anne R Meyer
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elena Kolobova
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Melinda A Engevik
- Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Janice A Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mitchell D Shub
- Phoenix Children's Hospital and University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | | | | | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
17
|
Wang S, Li X, Wang W, Zhang H, Xu S. Application of transcriptome analysis: Oxidative stress, inflammation and microtubule activity disorder caused by ammonia exposure may be the primary factors of intestinal microvilli deficiency in chicken. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 696:134035. [PMID: 31470328 DOI: 10.1016/j.scitotenv.2019.134035] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 06/10/2023]
Abstract
Ammonia (NH3), an inhaled harmful gas, is not only an important volatile in fertilizer production and ranching, but also the main basic component of haze. However, the effect and mechanism of NH3 on the intestines are still unclear. To investigate the intestinal toxicity of NH3 inhalation, morphological changes, transcriptome profiles and oxidative stress indicators of jejunum in broiler chicken exposed to NH3 for 42 days were examined. Results of morphological observation showed that NH3 exposure caused deficiency of jejunal microvilli and neutrophil infiltration. Transcriptomics sequencing identified 677 differential expressed genes (DEGs) including 358 up-regulated genes and 319 down-regulated genes. Enrichment analysis of obtained DEGs by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) found that biological functions and pathways affected by NH3 included antioxidant function, inflammation, microtubule and nutrition transport. Relative genes validation and chemical detection confirmed that NH3-induced oxidative stress by activating CYPs and inhibiting antioxidant enzymes promoted inflammatory response and decreased microtubule activity, thus destroying the balance of nutritional transporters. Our study perfects the injurious mechanism of NH3 exposure and provides a new insight and method for environmental risk assessment.
Collapse
Affiliation(s)
- Shengchen Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaojing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Wei Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
18
|
Jayawardena D, Alrefai WA, Dudeja PK, Gill RK. Recent advances in understanding and managing malabsorption: focus on microvillus inclusion disease. F1000Res 2019; 8. [PMID: 31824659 PMCID: PMC6896243 DOI: 10.12688/f1000research.20762.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2019] [Indexed: 12/11/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a rare congenital severe malabsorptive and secretory diarrheal disease characterized by blunted or absent microvilli with accumulation of secretory granules and inclusion bodies in enterocytes. The typical clinical presentation of the disease is severe chronic diarrhea that rapidly leads to dehydration and metabolic acidosis. Despite significant advances in our understanding of the causative factors, to date, no curative therapy for MVID and associated diarrhea exists. Prognosis mainly relies on life-long total parenteral nutrition (TPN) and eventual small bowel and/or liver transplantation. Both TPN and intestinal transplantation are challenging and present with many side effects. A breakthrough in the understanding of MVID emanated from seminal findings revealing mutations in
MYO5B as a cause for MVID. During the last decade, many studies have thus utilized cell lines and animal models with knockdown of
MYO5B to closely recapitulate the human disease and investigate potential therapeutic options in disease management. We will review the most recent advances made in the research pertaining to MVID. We will also highlight the tools and models developed that can be utilized for basic and applied research to increase our understanding of MVID and develop novel and effective targeted therapies.
Collapse
Affiliation(s)
- Dulari Jayawardena
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Waddah A Alrefai
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep K Dudeja
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Forteza R, Ahsan MK, Cartón-García F, Arango D, Ameen NA, Salas PJ. Glucocorticoids and myosin5b loss of function induce heightened PKA signaling in addition to membrane traffic defects. Mol Biol Cell 2019; 30:3076-3089. [PMID: 31664880 PMCID: PMC6938243 DOI: 10.1091/mbc.e18-07-0415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Loss-of-function mutations in the nonconventional myosin Vb (Myo5b) result in microvillus inclusion disease (MVID) and massive secretory diarrhea that often begins at birth. Myo5b mutations disrupt the apical recycling endosome (ARE) and membrane traffic, resulting in reduced surface expression of apical membrane proteins. ARE disruption also results in constitutive phosphoinositide-dependent kinase 1 gain of function. In MVID, decreased surface expression of apical anion channels involved in Cl- extrusion, such as cystic fibrosis transmembrane conductance regulator (CFTR), should reduce fluid secretion into the intestinal lumen. But the opposite phenotype is observed. To explain this contradiction and the onset of diarrhea, we hypothesized that signaling effects downstream from Myo5b loss of function synergize with higher levels of glucocorticoids to activate PKA and CFTR. Data from intestinal cell lines, human MVID, and Myo5b KO mouse intestine revealed changes in the subcellular redistribution of PKA activity to the apical pole, increased CFTR phosphorylation, and establishment of apical cAMP gradients in Myo5b-defective cells exposed to physiological levels of glucocorticoids. These cells also displayed net secretory fluid fluxes and transepithelial currents mainly from PKA-dependent Cl- secretion. We conclude that Myo5b defects result in PKA stimulation that activates residual channels on the surface when intestinal epithelia are exposed to glucocorticoids at birth.
Collapse
Affiliation(s)
- Radia Forteza
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - M Kaimul Ahsan
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06510
| | - Fernando Cartón-García
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autónoma de Barcelona, 08035 Barcelona, Spain
| | - Nadia A Ameen
- Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06510
| | - Pedro J Salas
- Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, FL 33136
| |
Collapse
|
20
|
Lee H, Kang J, Ahn S, Lee J. The Hippo Pathway Is Essential for Maintenance of Apicobasal Polarity in the Growing Intestine of Caenorhabditis elegans. Genetics 2019; 213:501-515. [PMID: 31358532 PMCID: PMC6781910 DOI: 10.1534/genetics.119.302477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022] Open
Abstract
Although multiple determinants for establishing polarity in membranes of epithelial cells have been identified, the mechanism for maintaining apicobasal polarity is not fully understood. Here, we show that the conserved Hippo kinase pathway plays a role in the maintenance of apicobasal polarity in the developing intestine of Caenorhabditis elegans We screened suppressors of the mutation in wts-1-the gene that encodes the LATS kinase homolog, deficiency of which leads to disturbance of the apicobasal polarity of the intestinal cells and to eventual death of the organism. We identified several alleles of yap-1 and egl-44 that suppress the effects of this mutation. yap-1 encodes a homolog of YAP/Yki, and egl-44 encodes a homolog of TEAD/Sd. WTS-1 bound directly to YAP-1 and inhibited its nuclear accumulation in intestinal cells. We also found that NFM-1, which is a homolog of NF2/Merlin, functioned in the same genetic pathway as WTS-1 to regulate YAP-1 to maintain cellular polarity. Transcriptome analysis identified several target candidates of the YAP-1-EGL-44 complex including TAT-2, which encodes a putative P-type ATPase. In summary, we have delineated the conserved Hippo pathway in C. elegans consisting of NFM-1-WTS-1-YAP-1-EGL-44 and proved that the proper regulation of YAP-1 by upstream NFM-1 and WTS-1 is essential for maintenance of apicobasal membrane identities of the growing intestine.
Collapse
Affiliation(s)
- Hanee Lee
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| | - Junsu Kang
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| | - Soungyub Ahn
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| | - Junho Lee
- Department of Biological Sciences, Seoul National University, Gwanak-gu 08826, Korea
| |
Collapse
|
21
|
Rao MC. Physiology of Electrolyte Transport in the Gut: Implications for Disease. Compr Physiol 2019; 9:947-1023. [PMID: 31187895 DOI: 10.1002/cphy.c180011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We now have an increased understanding of the genetics, cell biology, and physiology of electrolyte transport processes in the mammalian intestine, due to the availability of sophisticated methodologies ranging from genome wide association studies to CRISPR-CAS technology, stem cell-derived organoids, 3D microscopy, electron cryomicroscopy, single cell RNA sequencing, transgenic methodologies, and tools to manipulate cellular processes at a molecular level. This knowledge has simultaneously underscored the complexity of biological systems and the interdependence of multiple regulatory systems. In addition to the plethora of mammalian neurohumoral factors and their cross talk, advances in pyrosequencing and metagenomic analyses have highlighted the relevance of the microbiome to intestinal regulation. This article provides an overview of our current understanding of electrolyte transport processes in the small and large intestine, their regulation in health and how dysregulation at multiple levels can result in disease. Intestinal electrolyte transport is a balance of ion secretory and ion absorptive processes, all exquisitely dependent on the basolateral Na+ /K+ ATPase; when this balance goes awry, it can result in diarrhea or in constipation. The key transporters involved in secretion are the apical membrane Cl- channels and the basolateral Na+ -K+ -2Cl- cotransporter, NKCC1 and K+ channels. Absorption chiefly involves apical membrane Na+ /H+ exchangers and Cl- /HCO3 - exchangers in the small intestine and proximal colon and Na+ channels in the distal colon. Key examples of our current understanding of infectious, inflammatory, and genetic diarrheal diseases and of constipation are provided. © 2019 American Physiological Society. Compr Physiol 9:947-1023, 2019.
Collapse
Affiliation(s)
- Mrinalini C Rao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
22
|
Bidaud-Meynard A, Nicolle O, Heck M, Le Cunff Y, Michaux G. A V0-ATPase-dependent apical trafficking pathway maintains the polarity of the intestinal absorptive membrane. Development 2019; 146:dev174508. [PMID: 31110027 PMCID: PMC7376742 DOI: 10.1242/dev.174508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/07/2019] [Indexed: 12/18/2022]
Abstract
Intestine function relies on the strong polarity of intestinal epithelial cells and the array of microvilli forming a brush border at their luminal pole. Combining a genetic RNA interference (RNAi) screen with in vivo super-resolution imaging in the Caenorhabditiselegans intestine, we found that the V0 sector of the vacuolar ATPase (V0-ATPase) controls a late apical trafficking step, involving Ras-related protein 11 (RAB-11)+ endosomes and the N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE) synaptosome-associated protein 29 (SNAP-29), and is necessary to maintain the polarized localization of both apical polarity modules and brush border proteins. We show that the V0-ATPase pathway also genetically interacts with glycosphingolipids and clathrin in enterocyte polarity maintenance. Finally, we demonstrate that silencing of the V0-ATPase fully recapitulates the severe structural, polarity and trafficking defects observed in enterocytes from individuals with microvillus inclusion disease (MVID) and use this new in vivo MVID model to follow the dynamics of microvillus inclusions. Thus, we describe a new function for V0-ATPase in apical trafficking and epithelial polarity maintenance and the promising use of the C. elegans intestine as an in vivo model to better understand the molecular mechanisms of rare genetic enteropathies.
Collapse
Affiliation(s)
- Aurélien Bidaud-Meynard
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Ophélie Nicolle
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Markus Heck
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Yann Le Cunff
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Grégoire Michaux
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F-35000 Rennes, France
| |
Collapse
|
23
|
Bidaud-Meynard A, Bossard F, Schnúr A, Fukuda R, Veit G, Xu H, Lukacs GL. Transcytosis maintains CFTR apical polarity in the face of constitutive and mutation-induced basolateral missorting. J Cell Sci 2019; 132:jcs.226886. [PMID: 30975917 DOI: 10.1242/jcs.226886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 04/02/2019] [Indexed: 12/22/2022] Open
Abstract
Apical polarity of cystic fibrosis transmembrane conductance regulator (CFTR) is essential for solute and water transport in secretory epithelia and can be impaired in human diseases. Maintenance of apical polarity in the face of CFTR non-polarized delivery and inefficient apical retention of mutant CFTRs lacking PDZ-domain protein (NHERF1, also known as SLC9A3R1) interaction, remains enigmatic. Here, we show that basolateral CFTR delivery originates from biosynthetic (∼35%) and endocytic (∼65%) recycling missorting. Basolateral channels are retrieved via basolateral-to-apical transcytosis (hereafter denoted apical transcytosis), enhancing CFTR apical expression by two-fold and suppressing its degradation. In airway epithelia, CFTR transcytosis is microtubule-dependent but independent of Myo5B, Rab11 proteins and NHERF1 binding to its C-terminal DTRL motif. Increased basolateral delivery due to compromised apical recycling and accelerated internalization upon impaired NHERF1-CFTR association is largely counterbalanced by efficient CFTR basolateral internalization and apical transcytosis. Thus, transcytosis represents a previously unrecognized, but indispensable, mechanism for maintaining CFTR apical polarity that acts by attenuating its constitutive and mutation-induced basolateral missorting.
Collapse
Affiliation(s)
| | - Florian Bossard
- Department of Physiology, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Andrea Schnúr
- Department of Physiology, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Ryosuke Fukuda
- Department of Physiology, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Guido Veit
- Department of Physiology, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Haijin Xu
- Department of Physiology, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Gergely L Lukacs
- Department of Physiology, McGill University, Montréal, QC, H3G 1Y6, Canada .,Department of Biochemistry, McGill University, Montréal, QC, H3G 1Y6, Canada
| |
Collapse
|
24
|
Stephens CE, Whittamore JM, Hatch M. 125 Iodide as a surrogate tracer for epithelial chloride transport by the mouse large intestine in vitro. Exp Physiol 2019; 104:334-344. [PMID: 30615234 PMCID: PMC6397055 DOI: 10.1113/ep087445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022]
Abstract
NEW FINDINGS What is the central question of this study? The tracer 36 Cl- , currently used to measure transepithelial Cl- fluxes, has become prohibitively expensive, threatening its future use. 125 Iodide, previously validated alongside 36 Cl- as a tracer of Cl- efflux by cells, has not been tested as a surrogate for 36 Cl- across epithelia. What is the main finding and its importance? We demonstrate that 125 I- can serve as an inexpensive replacement for measuring Cl- transport across mouse large intestine, tracking Cl- transport in response to cAMP stimulation (inducing Cl- secretion) in the presence and absence of the main gastrointestinal Cl- -HCO3- exchanger, DRA. ABSTRACT Chloride transport is important for driving fluid secretion and absorption by the large intestine, with dysregulation resulting in diarrhoea-associated pathologies. The radioisotope 36 Cl- has long been used as a tracer to measure epithelial Cl- transport but is prohibitively expensive. 125 Iodide has been used as an alternative to 36 Cl- in some transport assays but has never been validated as an alternative for tracing bidirectional transepithelial Cl- fluxes. The goal of this study was to validate 125 I- as an alternative to 36 Cl- for measurement of Cl- transport by the intestine. Simultaneous fluxes of 36 Cl- and 125 I- were measured across the mouse caecum and distal colon. Net Cl- secretion was induced by the stimulation of cAMP with a cocktail of forskolin (FSK) and 3-isobutyl-1-methylxanthine (IBMX). Unidirectional fluxes of 125 I- correlated well with 36 Cl- fluxes after cAMP-induced net Cl- secretion, occurring predominantly through a reduction in the absorptive mucosal-to-serosal Cl- flux rather than by stimulation of the secretory serosal-to-mucosal Cl- flux. Correlations between 125 I- fluxes and 36 Cl- fluxes were maintained in epithelia from mice lacking DRA (Slc26a3), the main Cl- -HCO3- exchanger responsible for Cl- absorption by the large intestine. Lower rates of Cl- and I- absorption in the DRA knockout intestine suggest that DRA might have a previously unrecognized role in iodide uptake. This study validates that 125 I- traces transepithelial Cl- fluxes across the mouse large intestine, provides insights into the mechanism of net Cl- secretion and suggests that DRA might be involved in intestinal iodide absorption.
Collapse
Affiliation(s)
- Christine E Stephens
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jonathan M Whittamore
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marguerite Hatch
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
25
|
Engevik AC, Kaji I, Engevik MA, Meyer AR, Weis VG, Goldstein A, Hess MW, Müller T, Koepsell H, Dudeja PK, Tyska M, Huber LA, Shub MD, Ameen N, Goldenring JR. Loss of MYO5B Leads to Reductions in Na + Absorption With Maintenance of CFTR-Dependent Cl - Secretion in Enterocytes. Gastroenterology 2018; 155:1883-1897.e10. [PMID: 30144427 PMCID: PMC6279525 DOI: 10.1053/j.gastro.2018.08.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Inactivating mutations in MYO5B cause microvillus inclusion disease (MVID), but the physiological cause of the diarrhea associated with this disease is unclear. We investigated whether loss of MYO5B results in aberrant expression of apical enterocyte transporters. METHODS We studied alterations in apical membrane transporters in MYO5B-knockout mice, as well as mice with tamoxifen-inducible, intestine-specific disruption of Myo5b (VilCreERT2;Myo5bflox/flox mice) or those not given tamoxifen (controls). Intestinal tissues were collected from mice and analyzed by immunostaining, immunoelectron microscopy, or cultured enteroids were derived. Functions of brush border transporters in intestinal mucosa were measured in Ussing chambers. We obtained duodenal biopsy specimens from individuals with MVID and individuals without MVID (controls) and compared transporter distribution by immunocytochemistry. RESULTS Compared to intestinal tissues from littermate controls, intestinal tissues from MYO5B-knockout mice had decreased apical localization of SLC9A3 (also called NHE3), SLC5A1 (also called SGLT1), aquaporin (AQP) 7, and sucrase isomaltase, and subapical localization of intestinal alkaline phosphatase and CDC42. However, CFTR was present on apical membranes of enterocytes from MYO5B knockout and control mice. Intestinal biopsies from patients with MVID had subapical localization of NHE3, SGLT1, and AQP7, but maintained apical CFTR. After tamoxifen administration, VilCreERT2;Myo5bflox/flox mice lost apical NHE3, SGLT1, DRA, and AQP7, similar to germline MYO5B knockout mice. Intestinal tissues from VilCreERT2;Myo5bflox/flox mice had increased CFTR in crypts and CFTR localized to the apical membranes of enterocytes. Intestinal mucosa from VilCreERT2;Myo5bflox/flox mice given tamoxifen did not have an intestinal barrier defect, based on Ussing chamber analysis, but did have decreased SGLT1 activity and increased CFTR activity. CONCLUSIONS Although trafficking of many apical transporters is regulated by MYO5B, trafficking of CFTR is largely independent of MYO5B. Decreased apical localization of NHE3, SGLT1, DRA, and AQP7 might be responsible for dysfunctional water absorption in enterocytes of patients with MVID. Maintenance of apical CFTR might exacerbate water loss by active secretion of chloride into the intestinal lumen.
Collapse
Affiliation(s)
- Amy C Engevik
- Departments of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Izumi Kaji
- Departments of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Anne R Meyer
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Victoria G Weis
- Departments of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Anna Goldstein
- Departments of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | - Michael W Hess
- Division of Histology and Embryology, Innsbruck Medical University, Innsbruck, Austria
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Pradeep K Dudeja
- Department of Medicine, University of Illinois, Chicago and the Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Matthew Tyska
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lukas A Huber
- Division of Cell Biology, Biocenter and Innsbruck Medical University, Innsbruck, Austria; Austrian Drug Screening Institute, Innsbruck, Austria
| | - Mitchell D Shub
- Division of Gastroenterology and Phoenix Children's Hospital and the Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - Nadia Ameen
- Department of Pediatrics, Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - James R Goldenring
- Departments of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee.
| |
Collapse
|
26
|
Ahsan MK, Tchernychev B, Kessler MM, Solinga RM, Arthur D, Linde CI, Silos-Santiago I, Hannig G, Ameen NA. Linaclotide activates guanylate cyclase-C/cGMP/protein kinase-II-dependent trafficking of CFTR in the intestine. Physiol Rep 2018; 5:5/11/e13299. [PMID: 28592587 PMCID: PMC5471438 DOI: 10.14814/phy2.13299] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/10/2017] [Accepted: 05/03/2017] [Indexed: 12/12/2022] Open
Abstract
The transmembrane receptor guanylyl cyclase‐C (GC‐C), expressed on enterocytes along the intestine, is the molecular target of the GC‐C agonist peptide linaclotide, an FDA‐approved drug for treatment of adult patients with Irritable Bowel Syndrome with Constipation and Chronic Idiopathic Constipation. Polarized human colonic intestinal cells (T84, CaCo‐2BBe) rat and human intestinal tissues were employed to examine cellular signaling and cystic fibrosis transmembrane conductance regulator (CFTR)‐trafficking pathways activated by linaclotide using confocal microscopy, in vivo surface biotinylation, and protein kinase‐II (PKG‐II) activity assays. Expression and activity of GC‐C/cGMP pathway components were determined by PCR, western blot, and cGMP assays. Fluid secretion as a marker of CFTR cell surface translocation was determined using in vivo rat intestinal loops. Linaclotide treatment (30 min) induced robust fluid secretion and translocation of CFTR from subapical compartments to the cell surface in rat intestinal loops. Similarly, linaclotide treatment (30 min) of T84 and CaCo‐2BBe cells increased cell surface CFTR levels. Linaclotide‐induced activation of the GC‐C/cGMP/PKGII signaling pathway resulted in elevated intracellular cGMP and pVASPser239 phosphorylation. Inhibition or silencing of PKGII significantly attenuated linaclotide‐induced CFTR trafficking to the apical membrane. Inhibition of protein kinase‐A (PKA) also attenuated linaclotide‐induced CFTR cell surface trafficking, implying cGMP‐dependent cross‐activation of PKA pathway. Together, these findings support linaclotide‐induced activation of the GC‐C/cGMP/PKG‐II/CFTR pathway as the major pathway of linaclotide‐mediated intestinal fluid secretion, and that linaclotide‐dependent CFTR activation and recruitment/trafficking of CFTR from subapical vesicles to the cell surface is an important step in this process.
Collapse
Affiliation(s)
- Md Kaimul Ahsan
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut
| | - Boris Tchernychev
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | - Marco M Kessler
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | - Robert M Solinga
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | | | | | | | - Gerhard Hannig
- Department of Pharmacology, Ironwood Pharmaceuticals, Cambridge, Massachusetts
| | - Nadia A Ameen
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut .,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
27
|
Schneeberger K, Roth S, Nieuwenhuis EES, Middendorp S. Intestinal epithelial cell polarity defects in disease: lessons from microvillus inclusion disease. Dis Model Mech 2018; 11:11/2/dmm031088. [PMID: 29590640 PMCID: PMC5894939 DOI: 10.1242/dmm.031088] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium is a highly organized tissue. The establishment of epithelial cell polarity, with distinct apical and basolateral plasma membrane domains, is pivotal for both barrier formation and for the uptake and vectorial transport of nutrients. The establishment of cell polarity requires a specialized subcellular machinery to transport and recycle proteins to their appropriate location. In order to understand and treat polarity-associated diseases, it is necessary to understand epithelial cell-specific trafficking mechanisms. In this Review, we focus on cell polarity in the adult mammalian intestine. We discuss how intestinal epithelial polarity is established and maintained, and how disturbances in the trafficking machinery can lead to a polarity-associated disorder, microvillus inclusion disease (MVID). Furthermore, we discuss the recent developments in studying MVID, including the creation of genetically manipulated cell lines, mouse models and intestinal organoids, and their uses in basic and applied research. Summary: Microvillus inclusion disease serves as a useful model to enhance our understanding of the intestinal trafficking and polarity machinery in health and disease.
Collapse
Affiliation(s)
- Kerstin Schneeberger
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabrina Roth
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Edward E S Nieuwenhuis
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands
| | - Sabine Middendorp
- Division of Paediatrics, Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital, 3584 CT, Utrecht, The Netherlands .,Regenerative Medicine Center Utrecht, University Medical Centre (UMC) Utrecht, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
28
|
Dhekne HS, Pylypenko O, Overeem AW, Zibouche M, Ferreira RJ, van der Velde KJ, Rings EHHM, Posovszky C, van der Sluijs P, Swertz MA, Houdusse A, van IJzendoorn SCD. MYO5B, STX3, and STXBP2 mutations reveal a common disease mechanism that unifies a subset of congenital diarrheal disorders: A mutation update. Hum Mutat 2018; 39:333-344. [PMID: 29266534 PMCID: PMC5838515 DOI: 10.1002/humu.23386] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/15/2022]
Abstract
Microvillus inclusion disease (MVID) is a rare but fatal autosomal recessive congenital diarrheal disorder caused by MYO5B mutations. In 2013, we launched an open‐access registry for MVID patients and their MYO5B mutations (www.mvid-central.org). Since then, additional unique MYO5B mutations have been identified in MVID patients, but also in non‐MVID patients. Animal models have been generated that formally prove the causality between MYO5B and MVID. Importantly, mutations in two other genes, STXBP2 and STX3, have since been associated with variants of MVID, shedding new light on the pathogenesis of this congenital diarrheal disorder. Here, we review these additional genes and their mutations. Furthermore, we discuss recent data from cell studies that indicate that the three genes are functionally linked and, therefore, may constitute a common disease mechanism that unifies a subset of phenotypically linked congenital diarrheal disorders. We present new data based on patient material to support this. To congregate existing and future information on MVID geno‐/phenotypes, we have updated and expanded the MVID registry to include all currently known MVID‐associated gene mutations, their demonstrated or predicted functional consequences, and associated clinical information.
Collapse
Affiliation(s)
- Herschel S Dhekne
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Olena Pylypenko
- Structural Motility, Institute Curie, Centre de Reserche, Paris, France
| | - Arend W Overeem
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Malik Zibouche
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rosaria J Ferreira
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - K Joeri van der Velde
- Genomics Coordination Center, Department of Genetics, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Edmond H H M Rings
- Department of Pediatrics, Erasmus Medical Center Rotterdam, Erasmus University Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Carsten Posovszky
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Peter van der Sluijs
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, the Netherlands,Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Morris A Swertz
- Genomics Coordination Center, Department of Genetics, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Anne Houdusse
- Structural Motility, Institute Curie, Centre de Reserche, Paris, France
| | - Sven C D van IJzendoorn
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Engevik AC, Goldenring JR. Trafficking Ion Transporters to the Apical Membrane of Polarized Intestinal Enterocytes. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027979. [PMID: 28264818 DOI: 10.1101/cshperspect.a027979] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Epithelial cells lining the gastrointestinal tract require distinct apical and basolateral domains to function properly. Trafficking and insertion of enzymes and transporters into the apical brush border of intestinal epithelial cells is essential for effective digestion and absorption of nutrients. Specific critical ion transporters are delivered to the apical brush border to facilitate fluid and electrolyte uptake. Maintenance of these apical transporters requires both targeted delivery and regulated membrane recycling. Examination of altered apical trafficking in patients with Microvillus Inclusion disease caused by inactivating mutations in MYO5B has led to insights into the regulation of apical trafficking by elements of the apical recycling system. Modeling of MYO5B loss in cell culture and animal models has led to recognition of Rab11a and Rab8a as critical regulators of apical brush border function. All of these studies show the importance of apical membrane trafficking dynamics in maintenance of polarized epithelial cell function.
Collapse
Affiliation(s)
- Amy Christine Engevik
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232.,Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee 37232.,Nashville VA Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
30
|
Bridges RJ, Bradbury NA. Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator and Drugs: Insights from Cellular Trafficking. Handb Exp Pharmacol 2018; 245:385-425. [PMID: 29460152 DOI: 10.1007/164_2018_103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The eukaryotic cell is organized into membrane-delineated compartments that are characterized by specific cadres of proteins sustaining biochemically distinct cellular processes. The appropriate subcellular localization of proteins is key to proper organelle function and provides a physiological context for cellular processes. Disruption of normal trafficking pathways for proteins is seen in several genetic diseases, where a protein's absence for a specific subcellular compartment leads to organelle disruption, and in the context of an individual, a disruption of normal physiology. Importantly, several drug therapies can also alter protein trafficking, causing unwanted side effects. Thus, a deeper understanding of trafficking pathways needs to be appreciated as novel therapeutic modalities are proposed. Despite the promising efficacy of novel therapeutic agents, the intracellular bioavailability of these compounds has proved to be a potential barrier, leading to failures in treatments for various diseases and disorders. While endocytosis of drug moieties provides an efficient means of getting material into cells, the subsequent release and endosomal escape of materials into the cytosol where they need to act has been a barrier. An understanding of cellular protein/lipid trafficking pathways has opened up strategies for increasing drug bioavailability. Approaches to enhance endosomal exit have greatly increased the cytosolic bioavailability of drugs and will provide a means of investigating previous drugs that may have been shelved due to their low cytosolic concentration.
Collapse
Affiliation(s)
- Robert J Bridges
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA.
| |
Collapse
|
31
|
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: 5.6] [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
|
32
|
New Insights and Perspectives in Congenital Diarrheal Disorders. CURRENT PEDIATRICS REPORTS 2017. [DOI: 10.1007/s40124-017-0136-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
33
|
Vogel GF, Janecke AR, Krainer IM, Gutleben K, Witting B, Mitton SG, Mansour S, Ballauff A, Roland JT, Engevik AC, Cutz E, Müller T, Goldenring JR, Huber LA, Hess MW. Abnormal Rab11-Rab8-vesicles cluster in enterocytes of patients with microvillus inclusion disease. Traffic 2017; 18:453-464. [PMID: 28407399 DOI: 10.1111/tra.12486] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022]
Abstract
Microvillus inclusion disease (MVID) is a congenital enteropathy characterized by accumulation of vesiculo-tubular endomembranes in the subapical cytoplasm of enterocytes, historically termed "secretory granules." However, neither their identity nor pathophysiological significance is well defined. Using immunoelectron microscopy and tomography, we studied biopsies from MVID patients (3× Myosin 5b mutations and 1× Syntaxin3 mutation) and compared them to controls and genome-edited CaCo2 cell models, harboring relevant mutations. Duodenal biopsies from 2 patients with novel Myosin 5b mutations and typical clinical symptoms showed unusual ultrastructural phenotypes: aberrant subapical vesicles and tubules were prominent in the enterocytes, though other histological hallmarks of MVID were almost absent (ectopic intra-/intercellular microvilli, brush border atrophy). We identified these enigmatic vesiculo-tubular organelles as Rab11-Rab8-positive recycling compartments of altered size, shape and location harboring the apical SNARE Syntaxin3, apical transporters sodium-hydrogen exchanger 3 (NHE3) and cystic fibrosis transmembrane conductance regulator. Our data strongly indicate that in MVID disrupted trafficking between cargo vesicles and the apical plasma membrane is the primary cause of a defect of epithelial polarity and subsequent facultative loss of brush border integrity, leading to malabsorption. Furthermore, they support the notion that mislocalization of transporters, such as NHE3 substantially contributes to the reported sodium loss diarrhea.
Collapse
Affiliation(s)
- Georg F Vogel
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria.,Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Iris M Krainer
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Karin Gutleben
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Witting
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sahar Mansour
- Human Genetics Research Center, St. George's University of London, London, UK
| | | | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Amy C Engevik
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ernest Cutz
- Division of Pathology, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - James R Goldenring
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee.,Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lukas A Huber
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
34
|
Kumari V, Desai S, Ameen NA. AP2 α modulates cystic fibrosis transmembrane conductance regulator function in the human intestine. J Cyst Fibros 2017; 16:327-334. [PMID: 28438500 PMCID: PMC5502754 DOI: 10.1016/j.jcf.2017.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/15/2017] [Accepted: 03/21/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND AP2 is a clathrin-based endocytic adaptor complex comprising α, β2, μ2 and σ2 subunits. μ2 regulates CFTR endocytosis. The α subunit interacts with CFTR in the intestine but its physiologic significance is unclear. METHODS CFTR short circuit current was measured in intestinal T84 cells following shRNA knock down of AP2α (AP2αKD). Clathrin-coated structures (CCS) were immunolabeled and quantified in AP2αKD intestinal Caco2BBe (C2BBe) cells. GST tagged human AP2α appendage domain was cloned and its interaction with CFTR determined by GST pull down assay. RESULT AP2αKD in T84 cells resulted in higher CFTR current (57%) compared to control, consistent with increased functional CFTR and delayed endocytosis. Depletion of AP2α reduced CCS in C2BBe cells. Pull down assays revealed an interaction between human AP2α appendage domain and CFTR. CONCLUSION AP2 α interacts with and modulates CFTR function in the intestine by participating in clathrin assembly and recruitment of CFTR to CCS.
Collapse
Affiliation(s)
- Vandana Kumari
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, CT, United States
| | - Shruti Desai
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, CT, United States
| | - Nadia A Ameen
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, CT, United States; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|