1
|
Bowman DM, Meenderink LM, Thomas KS, Manning EH, Tyska MJ, Goldenring JR. Microvillus inclusion disease-causing MYO5B point mutations exert differential effects on motor function. J Biol Chem 2025; 301:108328. [PMID: 39978676 PMCID: PMC11964754 DOI: 10.1016/j.jbc.2025.108328] [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] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/12/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
Microvillus inclusion disease (MVID) is a rare congenital diarrheal disorder typically caused by loss of function mutations in the unconventional myosin, myosin 5b (MYO5B), which leads to the mistrafficking of apical components in enterocytes. MVID can manifest in two phenotypes: in both the intestine and liver or the liver alone. Although previous studies seeking to understand MVID disease pathology used MYO5B KO models, many patients have point mutations and thus express a dysfunctional MYO5B. How these point mutations lead to a broad spectrum of disease severity and the development of two distinct disease phenotypes is still not known. Here, we investigate the effect of MVID patient mutations on the function of the MYO5B motor domain, independent of cargo binding, using confocal imaging and fluorescence recovery after photobleaching. Patient mutations demonstrated a range of effects in these assays, from rigor-like behavior to loss of actin binding. Additionally, analysis of fluorescence recovery after photobleaching turnover kinetics suggests that some mutations negatively impact the ability of MYO5B to stay bound to actin. Collectively, our findings indicate that patient mutations affect the MYO5B motor domain in diverse ways, consistent with the spectrum of phenotypes observed in patients.
Collapse
Affiliation(s)
- Deanna M Bowman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Leslie M Meenderink
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA
| | - Kyra S Thomas
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Section of Surgical Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elizabeth H Manning
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA; Section of Surgical Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James R Goldenring
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Tennessee Valley Health Care System, Nashville, Tennessee, USA; Section of Surgical Science, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Kunii M, Harada A. Molecular mechanisms of polarized transport to the apical plasma membrane. Front Cell Dev Biol 2024; 12:1477173. [PMID: 39445332 PMCID: PMC11497131 DOI: 10.3389/fcell.2024.1477173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Cell polarity is essential for cellular function. Directional transport within a cell is called polarized transport, and it plays an important role in cell polarity. In this review, we will introduce the molecular mechanisms of polarized transport, particularly apical transport, and its physiological importance.
Collapse
Affiliation(s)
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, The University of Osaka, Osaka, Japan
| |
Collapse
|
3
|
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
|
4
|
Dopeso H, Rodrigues P, Cartón-García F, Macaya I, Bilic J, Anguita E, Jing L, Brotons B, Vivancos N, Beà L, Sánchez-Martín M, Landolfi S, Hernandez-Losa J, Ramon y Cajal S, Nieto R, Vicario M, Farre R, Schwartz S, van Ijzendoorn SC, Kobayashi K, Martinez-Barriocanal Á, Arango D. RhoA downregulation in the murine intestinal epithelium results in chronic Wnt activation and increased tumorigenesis. iScience 2024; 27:109400. [PMID: 38523777 PMCID: PMC10959657 DOI: 10.1016/j.isci.2024.109400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/23/2023] [Accepted: 02/28/2024] [Indexed: 03/26/2024] Open
Abstract
Rho GTPases are molecular switches regulating multiple cellular processes. To investigate the role of RhoA in normal intestinal physiology, we used a conditional mouse model overexpressing a dominant negative RhoA mutant (RhoAT19N) in the intestinal epithelium. Although RhoA inhibition did not cause an overt phenotype, increased levels of nuclear β-catenin were observed in the small intestinal epithelium of RhoAT19N mice, and the overexpression of multiple Wnt target genes revealed a chronic activation of Wnt signaling. Elevated Wnt signaling in RhoAT19N mice and intestinal organoids did not affect the proliferation of intestinal epithelial cells but significantly interfered with their differentiation. Importantly, 17-month-old RhoAT19N mice showed a significant increase in the number of spontaneous intestinal tumors. Altogether, our results indicate that RhoA regulates the differentiation of intestinal epithelial cells and inhibits tumor initiation, likely through the control of Wnt signaling, a key regulator of proliferation and differentiation in the intestine.
Collapse
Affiliation(s)
- Higinio Dopeso
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Paulo Rodrigues
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Fernando Cartón-García
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Irati Macaya
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Josipa Bilic
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Estefanía Anguita
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Li Jing
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Bruno Brotons
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Núria Vivancos
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Laia Beà
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Manuel Sánchez-Martín
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Servicio de Transgénesis, Nucleus, Universidad de Salamanca, 37007 Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Stefania Landolfi
- Translational Molecular Pathology, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Javier Hernandez-Losa
- Translational Molecular Pathology, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Santiago Ramon y Cajal
- Translational Molecular Pathology, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Rocío Nieto
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - María Vicario
- Digestive System Research Unit, Vall d’Hebron University Hospital Research Institute (VHIR), 08035 Barcelona, Spain
| | - Ricard Farre
- Department of Chronic Diseases and Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), Leuven 3000, Belgium
| | - Simo Schwartz
- Group of Drug Delivery and Targeting, Vall d'Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Clinical Biochemistry Department, Vall d'Hebron University Hospital, 08035 Barcelona, Spain
| | - Sven C.D. van Ijzendoorn
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, the Netherlands
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Águeda Martinez-Barriocanal
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, Vall d’Hebron University Hospital Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Group of Molecular Oncology, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
| |
Collapse
|
5
|
Fu L, Zou Y, Yu B, Hong D, Guan T, Hu J, Xu Y, Wu Y, Kou J, Lv Y. Background and roles: myosin in autoimmune diseases. Front Cell Dev Biol 2023; 11:1220672. [PMID: 37691828 PMCID: PMC10484797 DOI: 10.3389/fcell.2023.1220672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
The myosin superfamily is a group of molecular motors. Autoimmune diseases are characterized by dysregulation or deficiency of the immune tolerance mechanism, resulting in an immune response to the human body itself. The link between myosin and autoimmune diseases is much more complex than scientists had hoped. Myosin itself immunization can induce experimental autoimmune diseases of animals, and myosins were abnormally expressed in a number of autoimmune diseases. Additionally, myosin takes part in the pathological process of multiple sclerosis, Alzheimer's disease, Parkinson's disease, autoimmune myocarditis, myositis, hemopathy, inclusion body diseases, etc. However, research on myosin and its involvement in the occurrence and development of diseases is still in its infancy, and the underlying pathological mechanisms are not well understood. We can reasonably predict that myosin might play a role in new treatments of autoimmune diseases.
Collapse
Affiliation(s)
- Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yonghui Zou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi, China
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Jinfang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yi Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yaoqi Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi, China
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
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]
|
7
|
Dooley SA, Engevik KA, Digrazia J, Stubler R, Kaji I, Krystofiak E, Engevik AC. Myosin 5b is required for proper localization of the intermicrovillar adhesion complex in the intestinal brush border. Am J Physiol Gastrointest Liver Physiol 2022; 323:G501-G510. [PMID: 36218265 PMCID: PMC9639760 DOI: 10.1152/ajpgi.00212.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/27/2022] [Accepted: 10/03/2022] [Indexed: 01/31/2023]
Abstract
Intestinal enterocytes have an elaborate apical membrane of actin-rich protrusions known as microvilli. The organization of microvilli is orchestrated by the intermicrovillar adhesion complex (IMAC), which connects the distal tips of adjacent microvilli. The IMAC is composed of CDHR2 and CDHR5 as well as the scaffolding proteins USH1C, ANKS4B, and Myosin 7b (MYO7B). To create an IMAC, cells must transport the proteins to the apical membrane. Myosin 5b (MYO5B) is a molecular motor that traffics ion transporters to the apical membrane of enterocytes, and we hypothesized that MYO5B may also be responsible for the localization of IMAC proteins. To address this question, we used two different mouse models: 1) neonatal germline MYO5B knockout (MYO5B KO) mice and 2) adult intestinal-specific tamoxifen-inducible VillinCreERT2;MYO5Bflox/flox mice. In control mice, immunostaining revealed that CDHR2, CDHR5, USH1C, and MYO7B were highly enriched at the tips of the microvilli. In contrast, neonatal germline and adult MYO5B-deficient mice showed loss of apical CDHR2, CDHR5, and MYO7B in the brush border and accumulation in a subapical compartment. Colocalization analysis revealed decreased Mander's coefficients in adult inducible MYO5B-deficient mice compared with control mice for CDHR2, CDHR5, USH1C, and MYO7B. Scanning electron microscopy images further demonstrated aberrant microvilli packing in adult inducible MYO5B-deficient mouse small intestine. These data indicate that MYO5B is responsible for the delivery of IMAC components to the apical membrane.NEW & NOTEWORTHY The intestinal epithelium absorbs nutrients and water through an elaborate apical membrane of highly organized microvilli. Microvilli organization is regulated by the intermicrovillar adhesion complexes, which create links between neighboring microvilli and control microvilli packing and density. In this study, we report a new trafficking partner of the IMAC, Myosin 5b. Loss of Myosin 5b results in a disorganized brush border and failure of IMAC proteins to reach the distal tips of microvilli.
Collapse
Affiliation(s)
- Sarah A Dooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Jessica Digrazia
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Rachel Stubler
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Evan Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Amy C Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
8
|
Rathan-Kumar S, Roland JT, Momoh M, Goldstein A, Lapierre LA, Manning E, Mitchell L, Norman J, Kaji I, Goldenring JR. Rab11FIP1-deficient mice develop spontaneous inflammation and show increased susceptibility to colon damage. Am J Physiol Gastrointest Liver Physiol 2022; 323:G239-G254. [PMID: 35819177 PMCID: PMC9423785 DOI: 10.1152/ajpgi.00042.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
The small GTPase, Rab11a, regulates vesicle trafficking and cell polarity in epithelial cells through interaction with Rab11 family-interacting proteins (Rab11-FIPs). We hypothesized that deficiency of Rab11-FIP1 would affect mucosal integrity in the intestine. Global Rab11FIP1 knockout (KO) mice were generated by deletion of the second exon. Pathology of intestinal tissues was analyzed by immunostaining of colonic sections and RNA-sequencing of isolated colonic epithelial cells. A low concentration of dextran sodium sulfate (DSS, 2%) was added to drinking water for 5 days, and injury score was compared between Rab11FIP1 KO, Rab11FIP2 KO, and heterozygous littermates. Rab11FIP1 KO mice showed normal fertility and body weight gain. More frequent lymphoid patches and infiltration of macrophages and neutrophils were identified in Rab11FIP1 KO mice before the development of rectal prolapse compared with control mice. The population of trefoil factor 3 (TFF3)-positive goblet cells was significantly lower, and the ratio of proliferative to nonproliferative cells was higher in Rab11FIP1 KO colons. Transcription signatures indicated that Rab11FIP1 deletion downregulated genes that mediate stress tolerance response, whereas genes mediating the response to infection were significantly upregulated, consistent with the inflammatory responses in the steady state. Lack of Rab11FIP1 also resulted in abnormal accumulation of subapical vesicles in colonocytes and the internalization of transmembrane mucin, MUC13, with Rab14. After DSS treatment, Rab11FIP1 KO mice showed greater body weight loss and more severe mucosal damage than those in heterozygous littermates. These findings suggest that Rab11FIP1 is important for cytoprotection mechanisms and for the maintenance of colonic mucosal integrity.NEW & NOTEWORTHY Although Rab11FIP1 is important in membrane trafficking in epithelial cells, the gastrointestinal phenotype of Rab11FIP1 knockout (KO) mice had never been reported. This study demonstrated that Rab11FIP1 loss induces mistrafficking of Rab14 and MUC13 and decreases in colonic goblet cells, resulting in impaired mucosal integrity.
Collapse
Affiliation(s)
- Sudiksha Rathan-Kumar
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph T Roland
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Momoh
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna Goldstein
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne A Lapierre
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth Manning
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Louise Mitchell
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Jim Norman
- Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom
| | - Izumi Kaji
- Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - 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 School of Medicine, Nashville, Tennessee
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
9
|
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: 0.3] [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
|
10
|
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: 1.3] [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
|
11
|
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
|
12
|
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
|
13
|
Zheng Y, Peng Y, Zhang S, Zhao H, Chen W, Yang Y, Hu Z, Yin Q, Peng Y. Case Report: MYO5B Homozygous Variant c.2090+3A>T Causes Intron Retention Related to Chronic Cholestasis and Diarrhea. Front Genet 2022; 13:872836. [PMID: 35706451 PMCID: PMC9189387 DOI: 10.3389/fgene.2022.872836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Biallelically mutated MYO5B is associated with microvillus inclusion disease (MVID, MIM: 251850), cholestasis, or both. This study aims at validating the splicing alteration and clinical features of an intron variant for diagnosis.Case Presentation: A homozygous variant of MYO5B, NM_001080467.2:c.2090+3A > T (NP_001073936.1:p.?) in intron 17, was identified in a patient suffering from chronic cholestasis and diarrhea. Functional validation showed that this variant caused 185 bp of intron retention in its mRNA and was predicted to present a premature translation termination site for myoVb (p.Arg697fs*47) in the head motor domain. In addition, bowel biopsy revealed decreased microvilli and local lesions of microvillus inclusion in the duodena of the patient. The patient was presented with neonatal cholestasis leading to cirrhosis, intractable diarrhea, cholelithiasis, hepatic cyst, corneal opacity, and failure to thrive.Conclusion: Our study demonstrated an intronic homozygous variant of MYO5B that affected an intron, subsequently altering splicing and leading to combined cholestasis and MVID. Our results further supported the underlying genotype–phenotype correlations and extended clinical practices toward its diagnosis and management.
Collapse
Affiliation(s)
- Yu Zheng
- First Department of General Surgery & Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
| | - Yuming Peng
- First Department of General Surgery & Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
| | - Shuju Zhang
- First Department of General Surgery & Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
| | - Hongmei Zhao
- Department of Gastroenterology and Nutrition, Hunan Children’s Hospital, Changsha, China
| | - Weijian Chen
- Department of Pathology, Hunan Children’s Hospital, Changsha, China
| | - Yongjia Yang
- First Department of General Surgery & Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
| | - Zhengmao Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Qiang Yin
- First Department of General Surgery & Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
- *Correspondence: Qiang Yin, ; Yu Peng,
| | - Yu Peng
- First Department of General Surgery & Pediatrics Research Institute of Hunan Province, Hunan Children’s Hospital, Changsha, China
- *Correspondence: Qiang Yin, ; Yu Peng,
| |
Collapse
|
14
|
Wang L, Qiu YL, Xu HM, Zhu J, Li SJ, OuYang WX, Yang YF, Lu Y, Xie XB, Xing QH, Wang JS. MYO5B-associated diseases: Novel liver-related variants and genotype-phenotype correlation. Liver Int 2022; 42:402-411. [PMID: 34811877 DOI: 10.1111/liv.15104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Biallelic pathogenic variants in MYO5B cause microvillus inclusion disease (MVID), or familial intrahepatic cholestasis (FIC). The reported FIC patients are scarce and so the genotype-phenotype correlation has not been fully characterised. This study aimed to report more MYO5B-associated FIC patients and correlate genotypes to phenotypes in more detail. METHODS The phenotype and genetic data of 12 newly diagnosed MYO5B-associated (including 11 FIC) patients, as well as 118 previously reported patients with available genotypes, were summarised. Only patients with biallelic MYO5B variants were enrolled. Nonsense, frameshift, canonical splice sites, initiation codon loss, and single exon or multiexon deletion were defined as null MYO5B variants. RESULTS Phenotypically, 50 were isolated MVID, 47 involved both liver and intestine (combined), and 33 were isolated FIC (9 persistent, 15 recurrent, 3 transient, and 6 un-sub-classified) patients. The severity of intestinal manifestation was positively correlated to an increased number of null variants (ρ = 0.299, P = .001). All FIC patients carried at least one non-null variant, and the severity of cholestasis was correlated to the presence of a null variant (ρ = 0.420, P = .029). The proportion of FIC patients (16/29, 55%) harbouring missense/in-frame variants affecting the non-motor regions of MYO5B was significantly higher than that of MVID (3/25, 12%, P = .001) and combined patients (3/31, 10%, P = .000). 10 of the 29 FIC patients harboured missense/in-frame variants at the IQ motifs comparing to none in the 56 MVID and combined patients (P = .000). CONCLUSIONS The phenotype of MYO5B deficiency was associated with MYO5B genotypes, the nullity or the domain affected.
Collapse
Affiliation(s)
- Li Wang
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yi-Ling Qiu
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Hong-Mei Xu
- Department of Infectious Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Zhu
- Department of Infectious Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang-Jie Li
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| | - Wen-Xian OuYang
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| | - Yong-Feng Yang
- Department of Hepatology, The Second Hospital of Nanjing, Nanjing, China
| | - Yi Lu
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xin-Bao Xie
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Qing-He Xing
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jian-She Wang
- The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| |
Collapse
|
15
|
Advanced Microscopy for Liver and Gut Ultrastructural Pathology in Patients with MVID and PFIC Caused by MYO5B Mutations. J Clin Med 2021; 10:jcm10091901. [PMID: 33924896 PMCID: PMC8125609 DOI: 10.3390/jcm10091901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Mutations in the actin motor protein myosinVb (myo5b) cause aberrant apical cargo transport and the congenital enteropathy microvillus inclusion disease (MVID). Recently, missense mutations in myo5b were also associated with progressive familial intrahepatic cholestasis (MYO5B-PFIC). Here, we thoroughly characterized the ultrastructural and immuno-cytochemical phenotype of hepatocytes and duodenal enterocytes from a unique case of an adult MYO5B-PFIC patient who showed constant hepatopathy but only periodic enteric symptoms. Selected data from two other patients supported the findings. Advanced methods such as cryo-fixation, freeze-substitution, immuno-gold labeling, electron tomography and immuno-fluorescence microscopy complemented the standard procedures. Liver biopsies showed mislocalization of Rab11 and bile canalicular membrane proteins. Rab11-positive vesicles clustered around bile canaliculi and resembled subapical clusters of aberrant recycling endosomes in enterocytes from MVID patients. The adult patient studied in detail showed a severe, MVID-specific enterocyte phenotype, despite only a mild clinical intestinal presentation. This included mislocalization of numerous proteins essential for apical cargo transport and morphological alterations. We characterized the heterogeneous population of large catabolic organelles regarding their complex ultrastructure and differential distribution of autophagic and lysosomal marker proteins. Finally, we generated duodenal organoids/enteroids from biopsies that recapitulated all MVID hallmarks, demonstrating the potential of this disease model for personalized medicine.
Collapse
|
16
|
Mashukova A, Forteza R, Shah VN, Salas PJ. The cell polarity kinase Par1b/MARK2 activation selects specific NF-kB transcripts via phosphorylation of core mediator Med17/TRAP80. Mol Biol Cell 2021; 32:690-702. [PMID: 33596087 PMCID: PMC8108508 DOI: 10.1091/mbc.e20-10-0646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Par1b/MARK2 is a Ser/Thr kinase with pleiotropic effects that participates in the generation of apico-basal polarity in Caenorhabditis elegans. It is phosphorylated by atypical PKC(ι/λ) in Thr595 and inhibited. Because previous work showed a decrease in atypical protein kinase C (aPKC) activity under proinflammatory conditions, we analyzed the hypothesis that the resulting decrease in Thr595-MARK2 with increased kinase activity may also participate in innate immunity. We confirmed that pT595-MARK2 was decreased under inflammatory stimulation. The increase in MARK2 activity was verified by Par3 delocalization and rescue with a specific inhibitor. MARK2 overexpression significantly enhanced the transcriptional activity of NF-kB for a subset of transcripts. It also resulted in phosphorylation of a single band (∼Mr 80,000) coimmunoprecipitating with RelA, identified as Med17. In vitro phosphorylation showed direct phosphorylation of Med17 in Ser152 by recombinant MARK2. Expression of S152D-Med17 mimicked the effect of MARK2 activation on downstream transcriptional regulation, which was antagonized by S152A-Med17. The decrease in pThr595 phosphorylation was validated in aPKC-deficient mouse jejunal mucosae. The transcriptional effects were confirmed in transcriptome analysis and transcript enrichment determinations in cells expressing S152D-Med17. We conclude that theMARK2-Med17 axis represents a novel form of cross-talk between polarity signaling and transcriptional regulation including, but not restricted to, innate immunity responses.
Collapse
Affiliation(s)
- Anastasia Mashukova
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136.,Department of Medical Education, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314
| | - Radia Forteza
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Viraj N Shah
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Pedro J Salas
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
17
|
Redhai S, Boutros M. The Role of Organelles in Intestinal Function, Physiology, and Disease. Trends Cell Biol 2021; 31:485-499. [PMID: 33551307 DOI: 10.1016/j.tcb.2021.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
The intestine maintains homeostasis by coordinating internal biological processes to adjust to fluctuating external conditions. The intestinal epithelium is continuously renewed and comprises multiple cell types, including absorptive cells, secretory cells, and resident stem cells. An important feature of this organ is its ability to coordinate many processes including cell proliferation, differentiation, regeneration, damage/stress response, immune activity, feeding behavior, and age-related changes by using conserved signaling pathways. However, the subcellular spatial organization of these signaling events and the organelles involved has only recently been studied in detail. Here we discuss how organelles of intestinal cells serve to initiate, mediate, and terminate signals, that are vital for homeostasis.
Collapse
Affiliation(s)
- Siamak Redhai
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, BioQuant and Medical Faculty Mannheim, D-69120 Heidelberg, Germany.
| |
Collapse
|
18
|
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: 2.5] [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
|
19
|
Congenital Diarrhea and Cholestatic Liver Disease: Phenotypic Spectrum Associated with MYO5B Mutations. J Clin Med 2021; 10:jcm10030481. [PMID: 33525641 PMCID: PMC7865828 DOI: 10.3390/jcm10030481] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Myosin Vb (MYO5B) is a motor protein that facilitates protein trafficking and recycling in polarized cells by RAB11- and RAB8-dependent mechanisms. Biallelic MYO5B mutations are identified in the majority of patients with microvillus inclusion disease (MVID). MVID is an intractable diarrhea of infantile onset with characteristic histopathologic findings that requires life-long parenteral nutrition or intestinal transplantation. A large number of such patients eventually develop cholestatic liver disease. Bi-allelic MYO5B mutations are also identified in a subset of patients with predominant early-onset cholestatic liver disease. We present here the compilation of 114 patients with disease-causing MYO5B genotypes, including 44 novel patients as well as 35 novel MYO5B mutations, and an analysis of MYO5B mutations with regard to functional consequences. Our data support the concept that (1) a complete lack of MYO5B protein or early MYO5B truncation causes predominant intestinal disease (MYO5B-MVID), (2) the expression of full-length mutant MYO5B proteins with residual function causes predominant cholestatic liver disease (MYO5B-PFIC), and (3) the expression of mutant MYO5B proteins without residual function causes both intestinal and hepatic disease (MYO5B-MIXED). Genotype-phenotype data are deposited in the existing open MYO5B database in order to improve disease diagnosis, prognosis, and genetic counseling.
Collapse
|
20
|
Madrid RR, Mathews PD, Patta AC, Gonzales-Flores AP, Ramirez CA, Rigoni VL, Tavares-Dias M, Mertins O. Safety of oral administration of high doses of ivermectin by means of biocompatible polyelectrolytes formulation. Heliyon 2020; 7:e05820. [PMID: 33426351 PMCID: PMC7775035 DOI: 10.1016/j.heliyon.2020.e05820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/02/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
The FDA-approved drug ivermectin is applied for treatments of onchocerciasis and lymphatic filariasis. The anti-cancer and anti-viral activities have been demonstrated stressing possibilities for the drug repurposing and therefore new information on high dosage safety is on demand. We analyzed in vivo tissue responses for high doses of ivermectin using Corydoras fish as animal model. We made intestinal histology and hematologic assays after oral administration of ivermectin transported with polyelectrolytes formulation. Histology showed any apparent damage of intestinal tissues at 0.22–170 mg of ivermectin/kg body weight. Immunofluorescence evidenced delocalization of Myosin-Vb at enterocytes only for the higher dose. Hematology parameters showed random variations after 7 days from administration, but a later apparent recover after 14 and 21 days. The study evaluated the potential of high doses of oral administration of ivermectin formulation, which could be an alternative with benefits in high compliance therapies.
Collapse
Affiliation(s)
- Rafael R.M. Madrid
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, SP, Brazil
| | - Patrick D. Mathews
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, SP, Brazil
- Corresponding author.
| | - Ana C.M.F. Patta
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, SP, Brazil
| | - Anai P. Gonzales-Flores
- Post-Graduate Program in Tropical Biodiversity, Federal University of Amapá, 68903-419 Macapá, AP, Brazil
- Institute of Research of the Peruvian Amazon (IIAP, AQUAREC), 17000 Puerto Maldonado, Peru
| | - Carlos A.B. Ramirez
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, SP, Brazil
| | - Vera L.S. Rigoni
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, SP, Brazil
| | | | - Omar Mertins
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, SP, Brazil
- Corresponding author.
| |
Collapse
|
21
|
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: 0.6] [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
|
22
|
Abstract
Actin is a conserved cytoskeletal protein with essential functions. Here, we review the state-of-the-art reagents, tools and methods used to probe actin biology and functions in zebrafish embryo and larvae. We also discuss specific cell types and tissues where the study of actin in zebrafish has provided new insights into its functions.
Collapse
|
23
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
24
|
Overeem AW, Li Q, Qiu Y, Cartón‐García F, Leng C, Klappe K, Dronkers J, Hsiao N, Wang J, Arango D, van Ijzendoorn SC. A Molecular Mechanism Underlying Genotype-Specific Intrahepatic Cholestasis Resulting From MYO5B Mutations. Hepatology 2020; 72:213-229. [PMID: 31750554 PMCID: PMC7496772 DOI: 10.1002/hep.31002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/17/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Progressive familial intrahepatic cholestasis (PFIC) 6 has been associated with missense but not biallelic nonsense or frameshift mutations in MYO5B, encoding the motor protein myosin Vb (myoVb). This genotype-phenotype correlation and the mechanism through which MYO5B mutations give rise to PFIC are not understood. The aim of this study was to determine whether the loss of myoVb or expression of patient-specific myoVb mutants can be causally related to defects in canalicular protein localization and, if so, through which mechanism. APPROACH AND RESULTS We demonstrate that the cholestasis-associated substitution of the proline at amino acid position 600 in the myoVb protein to a leucine (P660L) caused the intracellular accumulation of bile canalicular proteins in vesicular compartments. Remarkably, the knockout of MYO5B in vitro and in vivo produced no canalicular localization defects. In contrast, the expression of myoVb mutants consisting of only the tail domain phenocopied the effects of the Myo5b-P660L mutation. Using additional myoVb and rab11a mutants, we demonstrate that motor domain-deficient myoVb inhibited the formation of specialized apical recycling endosomes and that its disrupting effect on the localization of canalicular proteins was dependent on its interaction with active rab11a and occurred at the trans-Golgi Network/recycling endosome interface. CONCLUSIONS Our results reveal a mechanism through which MYO5B motor domain mutations can cause the mislocalization of canalicular proteins in hepatocytes which, unexpectedly, does not involve myoVb loss-of-function but, as we propose, a rab11a-mediated gain-of-toxic function. The results explain why biallelic MYO5B mutations that affect the motor domain but not those that eliminate myoVb expression are associated with PFIC6.
Collapse
Affiliation(s)
- Arend W. Overeem
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Qinghong Li
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Yi‐Ling Qiu
- The Center for Pediatric Liver DiseasesChildren’s Hospital of Fudan UniversityShanghaiChina,Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Fernando Cartón‐García
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d’Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
| | - Changsen Leng
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Karin Klappe
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Just Dronkers
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Nai‐Hua Hsiao
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Jian‐She Wang
- The Center for Pediatric Liver DiseasesChildren’s Hospital of Fudan UniversityShanghaiChina,Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d’Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
| | - Sven C.D. van Ijzendoorn
- Department of Biomedical Sciences of Cells and SystemsSection Molecular Cell BiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| |
Collapse
|
25
|
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: 28] [Impact Index Per Article: 5.6] [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
|
26
|
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: 2.5] [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
|
27
|
Goldenring JR. The challenge of personalized cell biology: The example of microvillus inclusion disease. Traffic 2019; 21:169-171. [PMID: 31596022 DOI: 10.1111/tra.12703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 11/30/2022]
Abstract
Whole exome sequencing now provides a tool for rapid analysis of patients manifesting congenital diseases. Congenital diarrheal diseases provide a critical example of the challenges of combining identification of genetic mutations responsible for disease with characterization of the cell biological and cell physiological deficits observed in patients. Recent studies exploring the cellular events associated with loss of functional Myosin 5B (MYO5B) have demonstrated the importance of cell biological and physiological analyses to provide a greater understanding of the implications of pathological mutations. Development of enteroids derived from biopsies of patients with complex congenital diarrheal diseases provides a critical resource for evaluation of the cell biological impact of specific monogenic mutations on enterocyte function. The ability to identify putative causative mutations for congenital disease now provides an opportunity to coordinate the efforts of physicians and cell biologists in an effort to provide patients with personalized cell biology analysis to improve patient diagnosis and treatment.
Collapse
Affiliation(s)
- James R Goldenring
- Department of Surgery and Cell and Developmental Biology, and the Epithelial Biology Center, Vanderbilt University Medical Center, and the Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
28
|
Leng C, Overeem AW, Cartón-Garcia F, Li Q, Klappe K, Kuipers J, Cui Y, Zuhorn IS, Arango D, van IJzendoorn SCD. Loss of MYO5B expression deregulates late endosome size which hinders mitotic spindle orientation. PLoS Biol 2019; 17:e3000531. [PMID: 31682603 PMCID: PMC6855566 DOI: 10.1371/journal.pbio.3000531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/14/2019] [Accepted: 10/17/2019] [Indexed: 12/23/2022] Open
Abstract
Recycling endosomes regulate plasma membrane recycling. Recently, recycling endosome–associated proteins have been implicated in the positioning and orientation of the mitotic spindle and cytokinesis. Loss of MYO5B, encoding the recycling endosome–associated myosin Vb, is associated with tumor development and tissue architecture defects in the gastrointestinal tract. Whether loss of MYO5B expression affects mitosis is not known. Here, we demonstrate that loss of MYO5B expression delayed cytokinesis, perturbed mitotic spindle orientation, led to the misorientation of the plane of cell division during the course of mitosis, and resulted in the delamination of epithelial cells. Remarkably, the effects on spindle orientation, but not cytokinesis, were a direct consequence of physical hindrance by giant late endosomes, which were formed in a chloride channel–sensitive manner concomitant with a redistribution of chloride channels from the cell periphery to late endosomes upon loss of MYO5B. Rab7 availability was identified as a limiting factor for the development of giant late endosomes. In accordance, increasing rab7 availability corrected mitotic spindle misorientation and cell delamination in cells lacking MYO5B expression. In conclusion, we identified a novel role for MYO5B in the regulation of late endosome size control and identify the inability to control late endosome size as an unexpected novel mechanism underlying defects in cell division orientation and epithelial architecture. Loss of the recycling endosome-associated motor protein myosin Vb causes the formation of giant late endo-lysosomes; these in turn hinder the orientation of the mitotic spindle and chromosome segregation. Deregulated endosome size thus hampers faithful cell division.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arend W. Overeem
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Fernando Cartón-Garcia
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Qinghong Li
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Karin Klappe
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jeroen Kuipers
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Yingying Cui
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Sven C. D. van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
29
|
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: 1.5] [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
|
30
|
Engevik AC, Kaji I, Postema MM, Faust JJ, Meyer AR, Williams JA, Fitz GN, Tyska MJ, Wilson JM, Goldenring JR. Loss of myosin Vb promotes apical bulk endocytosis in neonatal enterocytes. J Cell Biol 2019; 218:3647-3662. [PMID: 31562230 PMCID: PMC6829668 DOI: 10.1083/jcb.201902063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/22/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022] Open
Abstract
In patients with inactivating mutations in myosin Vb (Myo5B), enterocytes show large inclusions lined by microvilli. The origin of inclusions in small-intestinal enterocytes in microvillus inclusion disease is currently unclear. We postulated that inclusions in Myo5b KO mouse enterocytes form through invagination of the apical brush border membrane. 70-kD FITC-dextran added apically to Myo5b KO intestinal explants accumulated in intracellular inclusions. Live imaging of Myo5b KO-derived enteroids confirmed the formation of inclusions from the apical membrane. Treatment of intestinal explants and enteroids with Dyngo resulted in accumulation of inclusions at the apical membrane. Inclusions in Myo5b KO enterocytes contained VAMP4 and Pacsin 2 (Syndapin 2). Myo5b;Pacsin 2 double-KO mice showed a significant decrease in inclusion formation. Our results suggest that apical bulk endocytosis in Myo5b KO enterocytes resembles activity-dependent bulk endocytosis, the primary mechanism for synaptic vesicle uptake during intense neuronal stimulation. Thus, apical bulk endocytosis mediates the formation of inclusions in neonatal Myo5b KO enterocytes.
Collapse
Affiliation(s)
- Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Izumi Kaji
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Meagan M Postema
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - James J Faust
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Anne R Meyer
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Janice A Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN
| | - Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, Bio5 Institute, University of Arizona, Tucson, AZ
| | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN .,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN.,The Epithelial Biology Center and Vanderbilt University School of Medicine, Nashville, TN.,The Nashville VA Medical Center, Nashville, TN
| |
Collapse
|
31
|
Grassini DR, da Silva J, Hall TE, Baillie GJ, Simons C, Parton RG, Hogan BM, Smith KA. Myosin Vb is required for correct trafficking of N-cadherin and cardiac chamber ballooning. Dev Dyn 2019; 248:284-295. [PMID: 30801852 DOI: 10.1002/dvdy.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND During heart morphogenesis, the cardiac chambers undergo ballooning: a process involving regionalized elongation of cardiomyocytes. Cardiomyocyte shape changes require reorganization of the actin cytoskeleton; however, the genetic regulation of this process is not well understood. RESULTS From a forward genetic screen, we identified the zebrafish uq 23ks mutant which manifests chamber ballooning defects. Whole-genome sequencing-mapping identified a truncating mutation in the gene, myo5b. myo5b encodes an atypical myosin required for endosome recycling and, consistent with this, increased vesicles were observed in myo5b mutant cardiomyocytes. Expression of RFP-Rab11a (a recycling endosome marker) confirmed increased recycling endosomes in cardiomyocytes of myo5b mutants. To investigate potential cargo of MyoVb-associated vesicles, we examined the adherens junction protein, N-cadherin. N-cadherin appeared mispatterned at cell junctions, and an increase in the number of intracellular particles was also apparent. Co-localization with RFP-Rab11a confirmed increased N-cadherin-positive recycling endosomes, demonstrating N-cadherin trafficking is perturbed in myo5b mutants. Finally, phalloidin staining showed disorganized F-actin in myo5b cardiomyocytes, suggesting the cytoskeleton fails to remodel, obstructing chamber ballooning. CONCLUSIONS MyoVb is required for cardiomyocyte endosomal recycling and appropriate N-cadherin localization during the onset of chamber ballooning. Cardiomyocytes lacking MyoVb are unable to reorganize their actin cytoskeleton, resulting in failed chamber ballooning. Developmental Dynamics 248:284-295, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniela R Grassini
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jason da Silva
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Gregory J Baillie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Cas Simons
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Benjamin M Hogan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kelly A Smith
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
32
|
Reynier M, Allart S, Goudounèche D, Moga A, Serre G, Simon M, Leprince C. The Actin-Based Motor Myosin Vb Is Crucial to Maintain Epidermal Barrier Integrity. J Invest Dermatol 2019; 139:1430-1438. [PMID: 30660668 DOI: 10.1016/j.jid.2018.12.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/05/2018] [Accepted: 12/12/2018] [Indexed: 01/07/2023]
Abstract
Myosin Vb (Myo5b) is an unconventional myosin involved in the actin-dependent transport and tethering of intracellular organelles. In the epidermis, granular keratinocytes accumulate cytoplasmic lamellar bodies (LBs), secretory vesicles released at the junction with the stratum corneum that participate actively in the maintenance of the epidermal barrier. We have previously demonstrated that LB biogenesis is controlled by the Rab11a guanosine triphosphate hydrolase, known for its ability to recruit the Myo5b motor. In order to better characterize the molecular pathway that controls LB trafficking, we analyzed the role of F-actin and Myo5b in the epidermis. We demonstrated that LB distribution in granular keratinocytes was dependent on a dynamic F-actin cytoskeleton. Myo5b was shown to be highly expressed in granular keratinocytes and associated with corneodesmosin-loaded LB. In reconstructed human epidermis, Myo5b silencing led to epidermal barrier defects associated with structural alterations of the stratum corneum and a reduced pool of LB showing signs of disordered maturation. Myo5b depletion also disturbed the expression and distribution of both LB cargoes and junctional components, such as claudin-1, which demonstrates its action on both LB trafficking and junctional complex composition. Together, our data reveal the essential role of Myo5b in maintaining the epidermal barrier integrity.
Collapse
Affiliation(s)
- Marie Reynier
- Unité Différenciation Epithéliale et Autoimmunité Rhumatoïde, U1056, Institut National de la Santé et de la Recherche Médicale, University of Toulouse, Toulouse, France
| | - Sophie Allart
- Centre de Physiopathologie de Toulouse Purpan, U1043, Institut National de la Santé et de la Recherche Médicale, TRI Genotoul, Toulouse, France
| | - Dominique Goudounèche
- Centre de Microscopie Electronique Appliquée à la Biologie, Faculté de Médecine Rangueil, University of Toulouse, Toulouse, France
| | | | - Guy Serre
- Unité Différenciation Epithéliale et Autoimmunité Rhumatoïde, U1056, Institut National de la Santé et de la Recherche Médicale, University of Toulouse, Toulouse, France
| | - Michel Simon
- Unité Différenciation Epithéliale et Autoimmunité Rhumatoïde, U1056, Institut National de la Santé et de la Recherche Médicale, University of Toulouse, Toulouse, France
| | - Corinne Leprince
- Unité Différenciation Epithéliale et Autoimmunité Rhumatoïde, U1056, Institut National de la Santé et de la Recherche Médicale, University of Toulouse, Toulouse, France.
| |
Collapse
|
33
|
Julia J, Shui V, Mittal N, Heim-Hall J, Blanco CL. Microvillus inclusion disease, a diagnosis to consider when abnormal stools and neurological impairments run together due to a rare syntaxin 3 gene mutation. J Neonatal Perinatal Med 2019; 12:313-319. [PMID: 30909251 DOI: 10.3233/npm-1852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
BACKGROUND Microvillus Inclusion Disease (MVID) was first described in the literature in 1978 with presentation of severe watery diarrhea, failure to thrive, and metabolic acidosis. Mutations in the myosin Vb (MYO5B) gene have been identified as causative for MVID, but other clinical manifestations and associations with novel mutations are lacking. METHODS We report a full-term infant admitted to the neonatal intensive care unit (NICU) with abdominal distension and inability to sustain full enteral feeds. A retrospective chart review and review of the literature was performed. RESULTS An infant with abnormal, mucoid-like stringy stools was incidentally found to have severe metabolic acidosis on routine lab monitoring. Acidosis corrected with total parenteral nutrition (TPN), but the infant experienced recurrent episodes of acidosis with enteral feeds. He was also noted to have abnormal ocular movements, fluctuating tonicity, and staring spells. He underwent an extensive workup and the diagnosis of microvillus inclusion disease was made by findings on electron microscopy. The diagnosis was confirmed with whole exome sequencing, showing a rare homozygous mutation in the syntaxin 3 (STX3) gene. This is the fifth reported patient with microvillus inclusion disease with a mutation in this gene, and the first with abnormal neurologic findings. CONCLUSION It is important to consider MVID in the differential diagnosis of a neonate or infant with abnormal stools, metabolic acidosis, with and without neurologic symptoms for prompt referral and treatment.
Collapse
Affiliation(s)
- Jacqueline Julia
- Department of Pediatrics, University of Texas Health Science Center at San Antonio and University Health System, San Antonio, TX, USA
| | - Valerie Shui
- Department of Pediatrics, University of Texas Health Science Center at San Antonio and University Health System, San Antonio, TX, USA
| | - Naveen Mittal
- Department of Pediatrics, University of Texas Health Science Center at San Antonio and University Health System, San Antonio, TX, USA
| | - Josefine Heim-Hall
- Department of Pathology, University of Texas Health Science Center at San Antonio and University Health System, San Antonio, TX, USA
| | - Cynthia L Blanco
- Department of Pediatrics, University of Texas Health Science Center at San Antonio and University Health System, San Antonio, TX, USA
| |
Collapse
|
34
|
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: 6.0] [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
|
35
|
Abstract
The delivery of intracellular material within cells is crucial for maintaining normal function. Myosins transport a wide variety of cargo, ranging from vesicles to ribonuclear protein particles (RNPs), in plants, fungi, and metazoa. The properties of a given myosin transporter are adapted to move on different actin filament tracks, either on the disordered actin networks at the cell cortex or along highly organized actin bundles to distribute their cargo in a localized manner or move it across long distances in the cell. Transport is controlled by selective recruitment of the myosin to its cargo that also plays a role in activation of the motor.
Collapse
Affiliation(s)
- Margaret A Titus
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
36
|
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: 53] [Impact Index Per Article: 7.6] [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
|
37
|
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: 6.0] [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
|
38
|
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: 2.6] [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
|
39
|
Klunder LJ, Faber KN, Dijkstra G, van IJzendoorn SCD. Mechanisms of Cell Polarity-Controlled Epithelial Homeostasis and Immunity in the Intestine. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027888. [PMID: 28213466 DOI: 10.1101/cshperspect.a027888] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intestinal epithelial cell polarity is instrumental to maintain epithelial homeostasis and balance communications between the gut lumen and bodily tissue, thereby controlling the defense against gastrointestinal pathogens and maintenance of immune tolerance to commensal bacteria. In this review, we highlight recent advances with regard to the molecular mechanisms of cell polarity-controlled epithelial homeostasis and immunity in the human intestine.
Collapse
Affiliation(s)
- Leon J Klunder
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Sven C D van IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| |
Collapse
|
40
|
Feng Q, Bonder EM, Engevik AC, Zhang L, Tyska MJ, Goldenring JR, Gao N. Disruption of Rab8a and Rab11a causes formation of basolateral microvilli in neonatal enteropathy. J Cell Sci 2017; 130:2491-2505. [PMID: 28596241 DOI: 10.1242/jcs.201897] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022] Open
Abstract
Misplaced formation of microvilli to basolateral domains and intracellular inclusions in enterocytes are pathognomonic features in congenital enteropathy associated with mutation of the apical plasma membrane receptor syntaxin 3 (STX3). Although the demonstrated binding of Myo5b to the Rab8a and Rab11a small GTPases in vitro implicates cytoskeleton-dependent membrane sorting, the mechanisms underlying the microvillar location defect remain unclear. By selective or combinatory disruption of Rab8a and Rab11a membrane traffic in vivo, we demonstrate that transport of distinct cargo to the apical brush border rely on either individual or both Rab regulators, whereas certain basolateral cargos are redundantly transported by both factors. Enterocyte-specific Rab8a and Rab11a double-knockout mouse neonates showed immediate postnatal lethality and more severe enteropathy than single knockouts, with extensive formation of microvilli along basolateral surfaces. Notably, following an inducible Rab11a deletion from neonatal enterocytes, basolateral microvilli were induced within 3 days. These data identify a potentially important and distinct mechanism for a characteristic microvillus defect exhibited by enterocytes of patients with neonatal enteropathy.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Amy C Engevik
- Department of Surgery, and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lanjing Zhang
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA.,Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08536, USA.,Rutgers Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08903, USA
| | - Matthew J Tyska
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - James R Goldenring
- Department of Surgery, and Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.,Nashville VA Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA .,Rutgers Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08903, USA
| |
Collapse
|
41
|
Qiu Y, Gong J, Feng J, Wang R, Han J, Liu T, Lu Y, Li L, Zhang M, Sheps JA, Wang N, Yan Y, Li J, Chen L, Borchers CH, Sipos B, Knisely A, Ling V, Xing Q, Wang J. Defects in myosin VB are associated with a spectrum of previously undiagnosed low γ-glutamyltransferase cholestasis. Hepatology 2017; 65:1655-1669. [PMID: 28027573 PMCID: PMC5413810 DOI: 10.1002/hep.29020] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 11/16/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022]
Abstract
Hereditary cholestasis in childhood and infancy with normal serum gamma-glutamyltransferase (GGT) activity is linked to several genes. Many patients, however, remain genetically undiagnosed. Defects in myosin VB (MYO5B; encoded by MYO5B) cause microvillus inclusion disease (MVID; MIM251850) with recurrent watery diarrhea. Cholestasis, reported as an atypical presentation in MVID, has been considered a side effect of parenteral alimentation. Here, however, we report on 10 patients who experienced cholestasis associated with biallelic, or suspected biallelic, mutations in MYO5B and who had neither recurrent diarrhea nor received parenteral alimentation. Seven of them are from two study cohorts, together comprising 31 undiagnosed low-GGT cholestasis patients; 3 are sporadic. Cholestasis in 2 patients was progressive, in 3 recurrent, in 2 transient, and in 3 uncategorized because of insufficient follow-up. Liver biopsy specimens revealed giant-cell change of hepatocytes and intralobular cholestasis with abnormal distribution of bile salt export pump (BSEP) at canaliculi, as well as coarse granular dislocation of MYO5B. Mass spectrometry of plasma demonstrated increased total bile acids, primary bile acids, and conjugated bile acids, with decreased free bile acids, similar to changes in BSEP-deficient patients. Literature review revealed that patients with biallelic mutations predicted to eliminate MYO5B expression were more frequent in typical MVID than in isolated-cholestasis patients (11 of 38 vs. 0 of 13). CONCLUSION MYO5B deficiency may underlie 20% of previously undiagnosed low-GGT cholestasis. MYO5B deficiency appears to impair targeting of BSEP to the canalicular membrane with hampered bile acid excretion, resulting in a spectrum of cholestasis without diarrhea. (Hepatology 2017;65:1655-1669).
Collapse
Affiliation(s)
- Yi‐Ling Qiu
- The Center for Pediatric Liver DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| | - Jing‐Yu Gong
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Jia‐Yan Feng
- Department of PathologyChildren's Hospital of Fudan UniversityShanghaiChina
| | | | - Jun Han
- University of Victoria−Genome BC Proteomics CentreUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Teng Liu
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Yi Lu
- The Center for Pediatric Liver DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| | - Li‐Ting Li
- The Center for Pediatric Liver DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| | - Mei‐Hong Zhang
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | | | - Neng‐Li Wang
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Yan‐Yan Yan
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Jia‐Qi Li
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
| | - Lian Chen
- Department of PathologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Christoph H. Borchers
- University of Victoria−Genome BC Proteomics CentreUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Bence Sipos
- Institute of General Pathology and NeuropathologyTübingen University HospitalTübingenGermany
| | - A.S. Knisely
- Institute of PathologyGraz Medical UniversityGrazAustria
| | - Victor Ling
- BC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Qing‐He Xing
- Institutes of Biomedical Sciences of Fudan UniversityShanghaiChina
| | - Jian‐She Wang
- Department of PediatricsJinshan Hospital of Fudan UniversityShanghaiChina
- Department of Infectious DiseasesChildren's Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
42
|
Vogel GF, Hess MW, Pfaller K, Huber LA, Janecke AR, Müller T. Towards understanding microvillus inclusion disease. Mol Cell Pediatr 2016; 3:3. [PMID: 26830108 PMCID: PMC4733813 DOI: 10.1186/s40348-016-0031-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/07/2016] [Indexed: 01/07/2023] Open
Abstract
Microvillus inclusion disease (MVID) is characterised by onset of intractable life-threatening watery diarrhoea during infancy. Transmission electron microscopy demonstrates shortening or absence of apical microvilli, pathognomonic microvillus inclusions in mature enterocytes and subapical accumulation of periodic acid-Schiff-positive granules or vesicles confirming diagnosis. Mutations in MYO5B have been found to cause MVID. In two patients with MVID, whole-exome sequencing of DNA revealed homozygous truncating mutations in STX3. Mutations in these genes disrupt trafficking between apical cargo vesicles and the apical plasma membrane. Thus, disturbed delivery of certain brush border membrane proteins is a common defect in MVID.
Collapse
Affiliation(s)
- Georg F Vogel
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kristian Pfaller
- Division of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| |
Collapse
|
43
|
Overeem AW, Posovszky C, Rings EHMM, Giepmans BNG, van IJzendoorn SCD. The role of enterocyte defects in the pathogenesis of congenital diarrheal disorders. Dis Model Mech 2016; 9:1-12. [PMID: 26747865 PMCID: PMC4728335 DOI: 10.1242/dmm.022269] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Congenital diarrheal disorders are rare, often fatal, diseases that are difficult to diagnose (often requiring biopsies) and that manifest in the first few weeks of life as chronic diarrhea and the malabsorption of nutrients. The etiology of congenital diarrheal disorders is diverse, but several are associated with defects in the predominant intestinal epithelial cell type, enterocytes. These particular congenital diarrheal disorders (CDDENT) include microvillus inclusion disease and congenital tufting enteropathy, and can feature in other diseases, such as hemophagocytic lymphohistiocytosis type 5 and trichohepatoenteric syndrome. Treatment options for most of these disorders are limited and an improved understanding of their molecular bases could help to drive the development of better therapies. Recently, mutations in genes that are involved in normal intestinal epithelial physiology have been associated with different CDDENT. Here, we review recent progress in understanding the cellular mechanisms of CDDENT. We highlight the potential of animal models and patient-specific stem-cell-based organoid cultures, as well as patient registries, to integrate basic and clinical research, with the aim of clarifying the pathogenesis of CDDENT and expediting the discovery of novel therapeutic strategies. Summary: Overview of the recent progress in our understanding of congenital diarrheal disorders, and the available models to study these diseases.
Collapse
Affiliation(s)
- Arend W Overeem
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Carsten Posovszky
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075 Ulm, Germany
| | - Edmond H M M Rings
- Department of Pediatrics, Erasmus Medical Center Rotterdam, Erasmus University Rotterdam, 3000 CB Rotterdam, The Netherlands Department of Pediatrics, Leiden University Medical Center, Leiden University, 2300 RC Leiden, The Netherlands
| | - Ben N G Giepmans
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Sven C D van IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
44
|
Sidhaye J, Pinto CS, Dharap S, Jacob T, Bhargava S, Sonawane M. The zebrafish goosepimples/myosin Vb mutant exhibits cellular attributes of human microvillus inclusion disease. Mech Dev 2016; 142:62-74. [PMID: 27497746 PMCID: PMC5161235 DOI: 10.1016/j.mod.2016.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022]
Abstract
Microvillus inclusion disease (MVID) is a life-threatening enteropathy characterised by malabsorption and incapacitating fluid loss due to chronic diarrhoea. Histological analysis has revealed that enterocytes in MVID patients exhibit reduction of microvilli, presence of microvillus inclusion bodies and intestinal villus atrophy, whereas genetic linkage analysis has identified mutations in myosin Vb gene as the main cause of MVID. In order to understand the cellular basis of MVID and the associated formation of inclusion bodies, an animal model that develops ex utero and is tractable genetically as well as by microscopy would be highly useful. Here we report that the intestine of the zebrafish goosepimples (gsp)/myosin Vb (myoVb) mutant shows severe reduction in intestinal folds - structures similar to mammalian villi. The loss of folds is further correlated with changes in the shape of enterocytes. In striking similarity with MVID patients, zebrafish gsp/myoVb mutant larvae exhibit microvillus atrophy, microvillus inclusions and accumulation of secretory material in enterocytes. We propose that the zebrafish gsp/myoVb mutant is a valuable model to study the pathophysiology of MVID. Furthermore, owing to the advantages of zebrafish in screening libraries of small molecules, the gsp mutant will be an ideal tool to identify compounds having therapeutic value against MVID. myosin Vb is expressed in the zebrafish intestine. goosepimples/myosin Vb function is essential for epithelial morphogenesis in the zebrafish intestine. The goosepimples mutant recapitulates pathognomonic features of microvillus inclusion disease. The function of myosin Vb in the intestine is conserved between fish and mammals.
Collapse
Affiliation(s)
- Jaydeep Sidhaye
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Clyde Savio Pinto
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Shweta Dharap
- Department of Biotechnology, Abasaheb Garware College, Pune, India
| | - Tressa Jacob
- Indian Institute of Science Education and Research, Pune, India
| | - Shobha Bhargava
- Department of Zoology, University of Pune, Ganeshkhind, Pune, India
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India.
| |
Collapse
|
45
|
Abstract
The vertebrate small intestine requires an enormous surface area to effectively absorb nutrients from food. Morphological adaptations required to establish this extensive surface include generation of an extremely long tube and convolution of the absorptive surface of the tube into villi and microvilli. In this Review, we discuss recent findings regarding the morphogenetic and molecular processes required for intestinal tube elongation and surface convolution, examine shared and unique aspects of these processes in different species, relate these processes to known human maladies that compromise absorptive function and highlight important questions for future research.
Collapse
Affiliation(s)
- Katherine D Walton
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrew M Freddo
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sha Wang
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Deborah L Gumucio
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
46
|
Kravtsov DV, Ahsan MK, Kumari V, van Ijzendoorn SCD, Reyes-Mugica M, Kumar A, Gujral T, Dudeja PK, Ameen NA. Identification of intestinal ion transport defects in microvillus inclusion disease. Am J Physiol Gastrointest Liver Physiol 2016; 311:G142-55. [PMID: 27229121 PMCID: PMC4967175 DOI: 10.1152/ajpgi.00041.2016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/08/2016] [Indexed: 01/31/2023]
Abstract
Loss of function mutations in the actin motor myosin Vb (Myo5b) lead to microvillus inclusion disease (MVID) and death in newborns and children. MVID results in secretory diarrhea, brush border (BB) defects, villus atrophy, and microvillus inclusions (MVIs) in enterocytes. How loss of Myo5b results in increased stool loss of chloride (Cl(-)) and sodium (Na(+)) is unknown. The present study used Myo5b loss-of-function human MVID intestine, polarized intestinal cell models of secretory crypt (T84) and villus resembling (CaCo2BBe, C2BBe) enterocytes lacking Myo5b in conjunction with immunofluorescence confocal stimulated emission depletion (gSTED) imaging, immunohistochemical staining, transmission electron microscopy, shRNA silencing, immunoblots, and electrophysiological approaches to examine the distribution, expression, and function of the major BB ion transporters NHE3 (Na(+)), CFTR (Cl(-)), and SLC26A3 (DRA) (Cl(-)/HCO3 (-)) that control intestinal fluid transport. We hypothesized that enterocyte maturation defects lead villus atrophy with immature secretory cryptlike enterocytes in the MVID epithelium. We investigated the role of Myo5b in enterocyte maturation. NHE3 and DRA localization and function were markedly reduced on the BB membrane of human MVID enterocytes and Myo5bKD C2BBe cells, while CFTR localization was preserved. Forskolin-stimulated CFTR ion transport in Myo5bKD T84 cells resembled that of control. Loss of Myo5b led to YAP1 nuclear retention, retarded enterocyte maturation, and a cryptlike phenotype. We conclude that preservation of functional CFTR in immature enterocytes, reduced functional expression of NHE3, and DRA contribute to Cl(-) and Na(+) stool loss in MVID diarrhea.
Collapse
Affiliation(s)
- Dmitri V. Kravtsov
- 1Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut;
| | - Md Kaimul Ahsan
- 1Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut;
| | - Vandana Kumari
- 1Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut;
| | - Sven C. D. van Ijzendoorn
- 2Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands;
| | | | - Anoop Kumar
- 4Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; and
| | - Tarunmeet Gujral
- 4Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; and
| | - Pradeep K. Dudeja
- 4Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois; and
| | - Nadia A. Ameen
- 1Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, New Haven, Connecticut; ,5Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
47
|
Forteza R, Figueroa Y, Mashukova A, Dulam V, Salas PJ. Conditional knockout of polarity complex (atypical) PKCι reveals an anti-inflammatory function mediated by NF-κB. Mol Biol Cell 2016; 27:2186-97. [PMID: 27226486 PMCID: PMC4945138 DOI: 10.1091/mbc.e16-02-0086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/20/2016] [Indexed: 01/27/2023] Open
Abstract
Atypical PKC, Par6, and Par3 constitute a conserved complex signaling cell asymmetry. In contrast to its role in other tissues, atypical PKC inhibits NF-κB activation in epithelia and may function in maintaining low levels of inflammation in addition to establishing apicobasal polarity. The conserved proteins of the polarity complex made up of atypical PKC (aPKC, isoforms ι and ζ), Par6, and Par3 determine asymmetry in several cell types, from Caenorhabditis elegans oocytes to vertebrate epithelia and neurons. We previously showed that aPKC is down-regulated in intestinal epithelia under inflammatory stimulation. Further, expression of constitutively active PKCι decreases NF-κB activity in an epithelial cell line, the opposite of the effect reported in other cells. Here we tested the hypothesis that aPKC has a dual function in epithelia, inhibiting the NF-κB pathway in addition to having a role in apicobasal polarity. We achieved full aPKC down-regulation in small intestine villi and colon surface epithelium using a conditional epithelium-specific knockout mouse. The results show that aPKC is dispensable for polarity after cell differentiation, except for known targets, including ROCK and ezrin, claudin-4 expression, and barrier permeability. The aPKC defect resulted in increased NF-κB activity, which could be rescued by IKK and ROCK inhibitors. It also increased expression of proinflammatory cytokines. In contrast, expression of anti-inflammatory IL-10 decreased. We conclude that epithelial aPKC acts upstream of multiple mechanisms that participate in the inflammatory response in the intestine, including, but not restricted to, NF-κB.
Collapse
Affiliation(s)
- Radia Forteza
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Yolanda Figueroa
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Anastasia Mashukova
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136 Department of Physiology, Nova Southeastern University, Ft. Lauderdale, FL 33314
| | - Vipin Dulam
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Pedro J Salas
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
48
|
Zhang X, Gao N. RAB and RHO GTPases regulate intestinal crypt cell homeostasis and enterocyte function. Small GTPases 2016; 7:59-64. [PMID: 27142493 DOI: 10.1080/21541248.2016.1159274] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent human and mouse genetic studies have highlighted important contributions of several small GTPases, in particular Rab8a, (1) Cdc42, (2-4) and Rab11a, (5-8) to the proper morphogenesis and function of the mature intestinal epithelia. Additional insights about the involvement of these factors in maintaining intestinal stem cell homeostasis have also been obtained. (9,10) These studies suggest a conserved vesicular and membrane trafficking program utilized by the gastrointestinal tissue to support the rapid epithelial cell turnover and the highly sophisticated physiology of mature epithelial cells.
Collapse
Affiliation(s)
- Xiao Zhang
- a Department of Biological Sciences , Rutgers University , Newark , NJ , USA
| | - Nan Gao
- a Department of Biological Sciences , Rutgers University , Newark , NJ , USA
| |
Collapse
|
49
|
Vogel GF, Klee KMC, Janecke AR, Müller T, Hess MW, Huber LA. Cargo-selective apical exocytosis in epithelial cells is conducted by Myo5B, Slp4a, Vamp7, and Syntaxin 3. J Cell Biol 2016; 211:587-604. [PMID: 26553929 PMCID: PMC4639860 DOI: 10.1083/jcb.201506112] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The motor protein Myo5B and t-SNARE Stx3 drive cargo-selective apical exocytosis in polarized epithelial cells in a pathway dependent on v-SNARE–like Slp4a, v-SNARE Vamp7, Sec1/Munc18-like protein Munc18-2, and the Rab11/8 cascade. Mutations in the motor protein Myosin Vb (Myo5B) or the soluble NSF attachment protein receptor Syntaxin 3 (Stx3) disturb epithelial polarity and cause microvillus inclusion disease (MVID), a lethal hereditary enteropathy affecting neonates. To understand the molecular mechanism of Myo5B and Stx3 interplay, we used genome editing to introduce a defined Myo5B patient mutation in a human epithelial cell line. Our results demonstrate a selective role of Myo5B and Stx3 for apical cargo exocytosis in polarized epithelial cells. Apical exocytosis of NHE3, CFTR (cystic fibrosis transmembrane conductance regulator), and GLUT5 required an interaction cascade of Rab11, Myo5B, Slp4a, Munc18-2, and Vamp7 with Stx3, which cooperate in the final steps of this selective apical traffic pathway. The brush border enzymes DPPIV and sucrase-isomaltase still correctly localize at the apical plasma membrane independent of this pathway. Hence, our work demonstrates how Myo5B, Stx3, Slp4a, Vamp7, Munc18-2, and Rab8/11 cooperate during selective apical cargo trafficking and exocytosis in epithelial cells and thereby provides further insight into MVID pathophysiology.
Collapse
Affiliation(s)
- Georg F Vogel
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria Division of Histology and Embryology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Katharina M C Klee
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria Institute of Molecular Biology, University of Innsbruck, 6020 Innsbruck, Austria Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020 Innsbruck, Austria
| | - Andreas R Janecke
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michael W Hess
- Division of Histology and Embryology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
50
|
Weis VG, Knowles BC, Choi E, Goldstein AE, Williams JA, Manning EH, Roland JT, Lapierre LA, Goldenring JR. Loss of MYO5B in mice recapitulates Microvillus Inclusion Disease and reveals an apical trafficking pathway distinct to neonatal duodenum. Cell Mol Gastroenterol Hepatol 2016; 2:131-157. [PMID: 27019864 PMCID: PMC4806369 DOI: 10.1016/j.jcmgh.2015.11.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/25/2015] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND AIMS Inactivating mutations in MYO5B cause severe neonatal diarrhea in Microvillus Inclusion Disease. Loss of active MYO5B causes the formation of pathognomonic inclusions and aberrations in brush border enzymes. METHODS We developed three mouse models of germline, constitutively intestinal targeted and inducible intestinal targeted deletion of MYO5B. The mice were evaluated for enterocyte cellular morphology. RESULTS Germline MYO5B KO mice showed early diarrhea and failure to thrive with evident microvillus inclusions and loss of apical transporters in the duodenum. IgG was present within inclusions. Apical transporters were lost and inclusions were present in the duodenum, but were nearly absent in the ileum. VillinCre;MYO5BF/F mice showed similar pathology and morphological changes in duodenal enterocytes. In contrast, when MYO5B KO was induced with tamoxifen treatment at 8 weeks of age, VillinCreERT2;MYO5BF/F mice developed severe diarrhea with loss of duodenal brush border enzymes, but few inclusions were observed in enterocytes. However, if tamoxifen is administered to 2-day-old VillinCreERT2;MYO5BF/F mice, prominent microvillus inclusions were observed. CONCLUSIONS The microvillus inclusions that develop after MYO5B loss reveal the presence of an unrecognized apical membrane trafficking pathway in neonatal duodenal enterocytes. However, the diarrheal pathology after MYO5B loss is due to deficits in transporter presentation at the apical membrane in duodenal enterocytes.
Collapse
Affiliation(s)
- Victoria G. Weis
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Byron C. Knowles
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Eunyoung Choi
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - Anna E. Goldstein
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Janice A. Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elizabeth H. Manning
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - Joseph T. Roland
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Lynne A. Lapierre
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| | - James R. Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|