1
|
Laan SNJ, Lenderink BG, Eikenboom JCJ, Bierings R. Endothelial colony-forming cells in the spotlight: insights into the pathophysiology of von Willebrand disease and rare bleeding disorders. J Thromb Haemost 2024; 22:3355-3365. [PMID: 39243860 DOI: 10.1016/j.jtha.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Endothelial cells deliver a vital contribution to the maintenance of hemostasis by constituting an anatomical as well as functional barrier between the blood and the rest of the body. Apart from the physical barrier function, endothelial cells maintain the hemostatic equilibrium by their pro- and anticoagulant functions. An important part of their procoagulant contribution is the production of von Willebrand factor (VWF), which is a carrier protein for coagulation factor VIII and facilitates the formation of a platelet plug. Thus, VWF is indispensable for both primary and secondary hemostasis, which is exemplified by the bleeding disorder von Willebrand disease that results from qualitative or quantitative deficiencies in VWF. A cellular model that was found to accurately reflect the endothelium and its secretory organelles are endothelial colony-forming cells, which can be readily isolated from peripheral blood and constitute a robust ex vivo model to investigate the donor's endothelial cell function. This review summarizes some of the valuable insights on biology of VWF and pathogenic mechanisms of von Willebrand disease that have been made possible using studies with endothelial colony-forming cells derived from patients with bleeding disorders.
Collapse
Affiliation(s)
- Sebastiaan N J Laan
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands; Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands. https://twitter.com/laan_bas
| | - Britte G Lenderink
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jeroen C J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
2
|
Dey H, Perez-Hurtado M, Heidelberger R. Syntaxin 3B: A SNARE Protein Required for Vision. Int J Mol Sci 2024; 25:10665. [PMID: 39408994 PMCID: PMC11476516 DOI: 10.3390/ijms251910665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Syntaxin 3 is a member of a large protein family of syntaxin proteins that mediate fusion between vesicles and their target membranes. Mutations in the ubiquitously expressed syntaxin 3A splice form give rise to a serious gastrointestinal disorder in humans called microvillus inclusion disorder, while mutations that additionally involve syntaxin 3B, a splice form that is expressed primarily in retinal photoreceptors and bipolar cells, additionally give rise to an early onset severe retinal dystrophy. In this review, we discuss recent studies elucidating the roles of syntaxin 3B and the regulation of syntaxin 3B functionality in membrane fusion and neurotransmitter release in the vertebrate retina.
Collapse
Affiliation(s)
| | | | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (H.D.)
| |
Collapse
|
3
|
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
|
4
|
Szabó L, Pollio AR, Vogel GF. Intracellular Trafficking Defects in Congenital Intestinal and Hepatic Diseases. Traffic 2024; 25:e12954. [PMID: 39187475 DOI: 10.1111/tra.12954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Enterocytes and liver cells fulfill important metabolic and barrier functions and are responsible for crucial vectorial secretive and absorptive processes. To date, genetic diseases affecting metabolic enzymes or transmembrane transporters in the intestine and the liver are better comprehended than mutations affecting intracellular trafficking. In this review, we explore the emerging knowledge on intracellular trafficking defects and their clinical manifestations in both the intestine and the liver. We provide a detailed overview including more investigated diseases such as the canonical, variant and associated forms of microvillus inclusion disease, as well as recently described pathologies, highlighting the complexity and disease relevance of several trafficking pathways. We give examples of how intracellular trafficking hubs, such as the apical recycling endosome system, the trans-Golgi network, lysosomes, or the Golgi-to-endoplasmic reticulum transport are involved in the pathomechanism and lead to disease. Ultimately, understanding these processes could spark novel therapeutic approaches, which would greatly improve the quality of life of the affected patients.
Collapse
Affiliation(s)
- Luca Szabó
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Adam R Pollio
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Friedrich Vogel
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
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
|
6
|
Avitzur Y, Jimenez L, Martincevic I, Acra S, Courtney-Martin G, Gray M, Hope K, Muise A, Prieto Jimenez PM, Taylor N, Thiagarajah JR, Martín MG. Diet management in congenital diarrheas and enteropathies - general concepts and disease-specific approach, a narrative review. Am J Clin Nutr 2024; 120:17-33. [PMID: 38734141 PMCID: PMC11251218 DOI: 10.1016/j.ajcnut.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Congenital diarrheas and enteropathies (CODE) are a group of rare, heterogenous, monogenic disorders that lead to chronic diarrhea in infancy. Definitive treatment is rarely available, and supportive treatment is the mainstay. Nutritional management in the form of either specialized formulas, restrictive diet, or parenteral nutrition support in CODE with poor enteral tolerance is the cornerstone of CODE treatment and long-term growth. The evidence to support the use of specific diet regimens and nutritional approaches in most CODE disorders is limited due to the rarity of these diseases and the scant published clinical experience. The goal of this review was to create a comprehensive guide for nutritional management in CODE, based on the currently available literature, disease mechanism, and the PediCODE group experience. Enteral diet management in CODE can be divided into 3 distinct conceptual frameworks: nutrient elimination, nutrient supplementation, and generalized nutrient restriction. Response to nutrient elimination or supplementation can lead to resolution or significant improvement in the chronic diarrhea of CODE and resumption of normal growth. This pattern can be seen in CODE due to carbohydrate malabsorption, defects in fat absorption, and occasionally in electrolyte transport defects. In contrast, general diet restriction is mainly supportive. However, occasionally it allows parenteral nutrition weaning or reduction over time, mainly in enteroendocrine defects and rarely in epithelial trafficking and polarity defects. Further research is required to better elucidate the role of diet in the treatment of CODE and the appropriate diet management for each disease.
Collapse
Affiliation(s)
- Yaron Avitzur
- Group for Improvement of Intestinal Function and Treatment (GIFT), Transplant and Regenerative Centre, SickKids Hospital, Toronto, ON, Canada; Division of Gastroenterology, Hepatology and Nutrition, SickKids Hospital, University of Toronto, Toronto, ON, Canada.
| | - Lissette Jimenez
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Congenital Enteropathy Program, Boston Children's Hospital, Boston, MA, United States;; Harvard Digestive Disease Center, Boston MA, United States
| | - Inez Martincevic
- Division of Gastroenterology, Hepatology and Nutrition, SickKids Hospital, University of Toronto, Toronto, ON, Canada
| | - Sari Acra
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Glenda Courtney-Martin
- Group for Improvement of Intestinal Function and Treatment (GIFT), Transplant and Regenerative Centre, SickKids Hospital, Toronto, ON, Canada; Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Megan Gray
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kayla Hope
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Aleixo Muise
- Division of Gastroenterology, Hepatology and Nutrition, SickKids Hospital, University of Toronto, Toronto, ON, Canada
| | - Paula M Prieto Jimenez
- Division of Gastroenterology and Nutrition, Department of Pediatrics, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Nancy Taylor
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Congenital Enteropathy Program, Boston Children's Hospital, Boston, MA, United States;; Harvard Digestive Disease Center, Boston MA, United States
| | - Martín G Martín
- Division of Gastroenterology and Nutrition, Department of Pediatrics, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
7
|
Pasquier N, Jaulin F, Peglion F. Inverted apicobasal polarity in health and disease. J Cell Sci 2024; 137:jcs261659. [PMID: 38465512 PMCID: PMC10984280 DOI: 10.1242/jcs.261659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Apicobasal epithelial polarity controls the functional properties of most organs. Thus, there has been extensive research on the molecular intricacies governing the establishment and maintenance of cell polarity. Whereas loss of apicobasal polarity is a well-documented phenomenon associated with multiple diseases, less is known regarding another type of apicobasal polarity alteration - the inversion of polarity. In this Review, we provide a unifying definition of inverted polarity and discuss multiple scenarios in mammalian systems and human health and disease in which apical and basolateral membrane domains are interchanged. This includes mammalian embryo implantation, monogenic diseases and dissemination of cancer cell clusters. For each example, the functional consequences of polarity inversion are assessed, revealing shared outcomes, including modifications in immune surveillance, altered drug sensitivity and changes in adhesions to neighboring cells. Finally, we highlight the molecular alterations associated with inverted apicobasal polarity and provide a molecular framework to connect these changes with the core cell polarity machinery and to explain roles of polarity inversion in health and disease. Based on the current state of the field, failure to respond to extracellular matrix (ECM) cues, increased cellular contractility and membrane trafficking defects are likely to account for most cases of inverted apicobasal polarity.
Collapse
Affiliation(s)
- Nicolas Pasquier
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
- Cell Adhesion and Cancer lab, University of Turku, FI-20520 Turku, Finland
| | - Fanny Jaulin
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
| | - Florent Peglion
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
| |
Collapse
|
8
|
Sun M, Pylypenko O, Zhou Z, Xu M, Li Q, Houdusse A, van IJzendoorn SCD. Uncovering the Relationship Between Genes and Phenotypes Beyond the Gut in Microvillus Inclusion Disease. Cell Mol Gastroenterol Hepatol 2024; 17:983-1005. [PMID: 38307491 PMCID: PMC11041842 DOI: 10.1016/j.jcmgh.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
Microvillus inclusion disease (MVID) is a rare condition that is present from birth and affects the digestive system. People with MVID experience severe diarrhea that is difficult to control, cannot absorb dietary nutrients, and struggle to grow and thrive. In addition, diverse clinical manifestations, some of which are life-threatening, have been reported in cases of MVID. MVID can be caused by variants in the MYO5B, STX3, STXBP2, or UNC45A gene. These genes produce proteins that have been functionally linked to each other in intestinal epithelial cells. MVID associated with STXBP2 variants presents in a subset of patients diagnosed with familial hemophagocytic lymphohistiocytosis type 5. MVID associated with UNC45A variants presents in most patients diagnosed with osteo-oto-hepato-enteric syndrome. Furthermore, variants in MYO5B or STX3 can also cause other diseases that are characterized by phenotypes that can co-occur in subsets of patients diagnosed with MVID. Recent studies involving clinical data and experiments with cells and animals revealed connections between specific phenotypes occurring outside of the digestive system and the type of gene variants that cause MVID. Here, we have reviewed these patterns and correlations, which are expected to be valuable for healthcare professionals in managing the disease and providing personalized care for patients and their families.
Collapse
Affiliation(s)
- Mingyue Sun
- Department of Biomedical Sciences of Cells and Systems, Center for Liver Digestive & Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Olena Pylypenko
- Dynamics of Intra-Cellular Organization, Institute Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Zhe Zhou
- Department of Biomedical Sciences of Cells and Systems, Center for Liver Digestive & Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mingqian Xu
- Department of Biomedical Sciences of Cells and Systems, Center for Liver Digestive & Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Qinghong Li
- Department of Biomedical Sciences of Cells and Systems, Center for Liver Digestive & Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anne Houdusse
- Structural Motility, Institute Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Sven C D van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, Center for Liver Digestive & Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
9
|
John EM, Sathyan S, Pournami F, Prithvi AK, Nandakumar A, Prabhakar J, Jain N. CODE Think! Rare Mutations of STX3 Causing Microvillus Inclusion Disease. J Pediatr Genet 2023; 12:352. [PMID: 38162158 PMCID: PMC10756714 DOI: 10.1055/s-0043-1772207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/11/2023] [Indexed: 01/03/2024]
Affiliation(s)
- Elizabeth Mary John
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Sajina Sathyan
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Femitha Pournami
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Ajai Kumar Prithvi
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Anand Nandakumar
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Jyothi Prabhakar
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Naveen Jain
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| |
Collapse
|
10
|
Poplaski V, Bomidi C, Kambal A, Nguyen-Phuc H, Di Rienzi SC, Danhof HA, Zeng XL, Feagins LA, Deng N, Vilar E, McAllister F, Coarfa C, Min S, Kim HJ, Shukla R, Britton R, Estes MK, Blutt SE. Human intestinal organoids from Cronkhite-Canada syndrome patients reveal link between serotonin and proliferation. J Clin Invest 2023; 133:e166884. [PMID: 37909332 PMCID: PMC10617781 DOI: 10.1172/jci166884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 08/29/2023] [Indexed: 11/03/2023] Open
Abstract
Cronkhite-Canada Syndrome (CCS) is a rare, noninherited polyposis syndrome affecting 1 in every million individuals. Despite over 50 years of CCS cases, the etiopathogenesis and optimal treatment for CCS remains unknown due to the rarity of the disease and lack of model systems. To better understand the etiology of CCS, we generated human intestinal organoids (HIOs) from intestinal stem cells isolated from 2 patients. We discovered that CCS HIOs are highly proliferative and have increased numbers of enteroendocrine cells producing serotonin (also known as 5-hydroxytryptamine or 5HT). These features were also confirmed in patient tissue biopsies. Recombinant 5HT increased proliferation of non-CCS donor HIOs and inhibition of 5HT production in the CCS HIOs resulted in decreased proliferation, suggesting a link between local epithelial 5HT production and control of epithelial stem cell proliferation. This link was confirmed in genetically engineered HIOs with an increased number of enteroendocrine cells. This work provides a new mechanism to explain the pathogenesis of CCS and illustrates the important contribution of HIO cultures to understanding disease etiology and in the identification of novel therapies. Our work demonstrates the principle of using organoids for personalized medicine and sheds light on how intestinal hormones can play a role in intestinal epithelial proliferation.
Collapse
Affiliation(s)
- Victoria Poplaski
- Program in Translational Biology and Molecular Medicine
- Department of Molecular Virology and Microbiology, and
| | | | - Amal Kambal
- Department of Molecular Virology and Microbiology, and
| | | | - Sara C. Di Rienzi
- Department of Molecular Virology and Microbiology, and
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Heather A. Danhof
- Department of Molecular Virology and Microbiology, and
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Xi-Lei Zeng
- Department of Molecular Virology and Microbiology, and
| | - Linda A. Feagins
- Department of Internal Medicine, Center for Inflammatory Bowl Diseases, The University of Texas at Austin Dell Medical School, Austin, Texas, USA
| | - Nan Deng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston Texas, USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston Texas, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston Texas, USA
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center and
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Soyoun Min
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hyun Jung Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richa Shukla
- Department of Medicine, Section of Gasteroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert Britton
- Department of Molecular Virology and Microbiology, and
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, and
- Department of Medicine, Section of Gasteroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston Texas, USA
| | | |
Collapse
|
11
|
Nishad R, Betancourt-Solis M, Dey H, Heidelberger R, McNew JA. Regulation of Syntaxin3B-Mediated Membrane Fusion by T14, Munc18, and Complexin. Biomolecules 2023; 13:1463. [PMID: 37892145 PMCID: PMC10604575 DOI: 10.3390/biom13101463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Retinal neurons that form ribbon-style synapses operate over a wide dynamic range, continuously relaying visual information to their downstream targets. The remarkable signaling abilities of these neurons are supported by specialized presynaptic machinery, one component of which is syntaxin3B. Syntaxin3B is an essential t-SNARE protein of photoreceptors and bipolar cells that is required for neurotransmitter release. It has a light-regulated phosphorylation site in its N-terminal domain at T14 that has been proposed to modulate membrane fusion. However, a direct test of the latter has been lacking. Using a well-controlled in vitro fusion assay, we found that a phosphomimetic T14 syntaxin3B mutation leads to a small but significant enhancement of SNARE-mediated membrane fusion following the formation of the t-SNARE complex. While the addition of Munc18a had only a minimal effect on membrane fusion mediated by SNARE complexes containing wild-type syntaxin3B, a more significant enhancement was observed in the presence of Munc18a when the SNARE complexes contained a syntaxin3B T14 phosphomimetic mutant. Finally, we showed that the retinal-specific complexins (Cpx III and Cpx IV) inhibited membrane fusion mediated by syntaxin3B-containing SNARE complexes in a dose-dependent manner. Collectively, our results establish that membrane fusion mediated by syntaxin3B-containing SNARE complexes is regulated by the T14 residue of syntaxin3B, Munc18a, and Cpxs III and IV.
Collapse
Affiliation(s)
- Rajkishor Nishad
- Department of BioSciences, Rice University, 6500 Main Street, MS 601, Houston, TX 77005, USA;
| | - Miguel Betancourt-Solis
- Department of BioSciences, Rice University, 6500 Main Street, MS 601, Houston, TX 77005, USA;
- Lonza Biologics, 14905 Kirby Dr, Houston, TX 77047, USA
| | - Himani Dey
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center, Houston (UTHealth Houston), 6431 Fannin Street, Houston, TX 77030, USA;
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center, Houston (UTHealth Houston), 6431 Fannin Street, Houston, TX 77030, USA;
| | - James A. McNew
- Department of BioSciences, Rice University, 6500 Main Street, MS 601, Houston, TX 77005, USA;
| |
Collapse
|
12
|
Perez-Hurtado M, Dao C, Saenz AE, Heidelberger R. Syntaxin 3 is haplosufficient for long-term photoreceptor survival in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1208805. [PMID: 37609371 PMCID: PMC10443939 DOI: 10.3389/fopht.2023.1208805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Biallelic loss-of-function mutations in the syntaxin 3 gene have been linked to a severe retinal dystrophy in humans that presents in early childhood. In mouse models, biallelic inactivation of the syntaxin 3 gene in photoreceptors rapidly leads to their death. What is not known is whether a monoallelic syntaxin 3 loss-of-function mutation might cause photoreceptor loss with advancing age. To address this question, we compared the outer nuclear layer of older adult mice (≈ 20 months of age) that were heterozygous for syntaxin 3 with those of similarly-aged control mice. We found that the photoreceptor layer maintains its thickness in mice that are heterozygous for syntaxin 3 relative to controls and that photoreceptor somatic counts are comparable. In addition, dendritic sprouting of the rod bipolar cell dendrites into the outer nuclear layer, which occurs following the loss of functional rod targets, was similar between genotypes. Thus, syntaxin 3 appears to be haplosufficient for photoreceptor survival, even with advancing age.
Collapse
Affiliation(s)
- Mariajose Perez-Hurtado
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
| | - Calvin Dao
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
| | - Amanda E. Saenz
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX
- The University of Texas MDAnderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston TX
| |
Collapse
|
13
|
Babcock SJ, Flores-Marin D, Thiagarajah JR. The genetics of monogenic intestinal epithelial disorders. Hum Genet 2023; 142:613-654. [PMID: 36422736 PMCID: PMC10182130 DOI: 10.1007/s00439-022-02501-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/23/2022] [Indexed: 11/27/2022]
Abstract
Monogenic intestinal epithelial disorders, also known as congenital diarrheas and enteropathies (CoDEs), are a group of rare diseases that result from mutations in genes that primarily affect intestinal epithelial cell function. Patients with CoDE disorders generally present with infantile-onset diarrhea and poor growth, and often require intensive fluid and nutritional management. CoDE disorders can be classified into several categories that relate to broad areas of epithelial function, structure, and development. The advent of accessible and low-cost genetic sequencing has accelerated discovery in the field with over 45 different genes now associated with CoDE disorders. Despite this increasing knowledge in the causal genetics of disease, the underlying cellular pathophysiology remains incompletely understood for many disorders. Consequently, clinical management options for CoDE disorders are currently limited and there is an urgent need for new and disorder-specific therapies. In this review, we provide a general overview of CoDE disorders, including a historical perspective of the field and relationship to other monogenic disorders of the intestine. We describe the genetics, clinical presentation, and known pathophysiology for specific disorders. Lastly, we describe the major challenges relating to CoDE disorders, briefly outline key areas that need further study, and provide a perspective on the future genetic and therapeutic landscape.
Collapse
Affiliation(s)
- Stephen J Babcock
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA
| | - David Flores-Marin
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Patrício D, Santiago J, Mano JF, Fardilha M. Organoids of the male reproductive system: Challenges, opportunities, and their potential use in fertility research. WIREs Mech Dis 2023; 15:e1590. [PMID: 36442887 DOI: 10.1002/wsbm.1590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/17/2022] [Accepted: 11/12/2022] [Indexed: 11/30/2022]
Abstract
Organoids are units of function of a given organ able to reproduce, in culture, a biological structure similar in architecture and function to its counterpart in vivo. Today, it is possible to develop an organoid from a fragment of tissue, a stem cell located in an adult organ, an embryonic stem cell, or an induced pluripotent stem cell. In the past decade, many organoids have been developed which mimic stomach, pancreas, liver and brain tissues, optic cups, among many others. Additionally, different male reproductive system organs have already been developed as organoids, including the prostate and testis. These 3D cultures may be of great importance for urological cancer research and have the potential to be used in fertility research for the study of spermatozoa production and maturation, germ cells-somatic cells interactions, and mechanisms of disease. They also provide an accurate preclinical pipeline for drug testing and discovery, as well as for the study of drug resistance. In this work, we revise the current knowledge on organoid technology and its use in healthcare and research, describe the male reproductive system organoids and other biomaterials already developed, and discuss their current application. Finally, we highlight the research gaps, challenges, and opportunities in the field and propose strategies to improve the use of organoids for the study of male infertility situations. This article is categorized under: Reproductive System Diseases > Stem Cells and Development Reproductive System Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Daniela Patrício
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.,Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Joana Santiago
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Margarida Fardilha
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
15
|
Klee KMC, Hess MW, Lohmüller M, Herzog S, Pfaller K, Müller T, Vogel GF, Huber LA. A CRISPR screen in intestinal epithelial cells identifies novel factors for polarity and apical transport. eLife 2023; 12:e80135. [PMID: 36661306 PMCID: PMC9889089 DOI: 10.7554/elife.80135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 01/19/2023] [Indexed: 01/21/2023] Open
Abstract
Epithelial polarization and polarized cargo transport are highly coordinated and interdependent processes. In our search for novel regulators of epithelial polarization and protein secretion, we used a genome-wide CRISPR/Cas9 screen and combined it with an assay based on fluorescence-activated cell sorting (FACS) to measure the secretion of the apical brush-border hydrolase dipeptidyl peptidase 4 (DPP4). In this way, we performed the first CRISPR screen to date in human polarized epithelial cells. Using high-resolution microscopy, we detected polarization defects and mislocalization of DPP4 to late endosomes/lysosomes after knockout of TM9SF4, anoctamin 8, and ARHGAP33, confirming the identification of novel factors for epithelial polarization and apical cargo secretion. Thus, we provide a powerful tool suitable for studying polarization and cargo secretion in epithelial cells. In addition, we provide a dataset that serves as a resource for the study of novel mechanisms for epithelial polarization and polarized transport and facilitates the investigation of novel congenital diseases associated with these processes.
Collapse
Affiliation(s)
- Katharina MC Klee
- Institute of Cell Biology, Medical University of InnsbruckInnsbruckAustria
- Institute of Histology and Embryology, Medical University of InnsbruckInnsbruckAustria
| | - Michael W Hess
- Institute of Histology and Embryology, Medical University of InnsbruckInnsbruckAustria
| | - Michael Lohmüller
- Institute of Developmental Immunology, Medical University of InnsbruckInnsbruckAustria
| | - Sebastian Herzog
- Institute of Developmental Immunology, Medical University of InnsbruckInnsbruckAustria
| | - Kristian Pfaller
- Institute of Histology and Embryology, Medical University of InnsbruckInnsbruckAustria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of InnsbruckInnsbruckAustria
| | - Georg F Vogel
- Institute of Cell Biology, Medical University of InnsbruckInnsbruckAustria
- Department of Paediatrics I, Medical University of InnsbruckInnsbruckAustria
| | - Lukas A Huber
- Institute of Cell Biology, Medical University of InnsbruckInnsbruckAustria
| |
Collapse
|
16
|
Novelli G, Spitalieri P, Murdocca M, Centanini E, Sangiuolo F. Organoid factory: The recent role of the human induced pluripotent stem cells (hiPSCs) in precision medicine. Front Cell Dev Biol 2023; 10:1059579. [PMID: 36699015 PMCID: PMC9869172 DOI: 10.3389/fcell.2022.1059579] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
During the last decades, hiPSC-derived organoids have been extensively studied and used as in vitro models for several applications among which research studies. They can be considered as organ and tissue prototypes, especially for those difficult to obtain. Moreover, several diseases can be accurately modeled and studied. Hence, patient-derived organoids (PDOs) can be used to predict individual drug responses, thus paving the way toward personalized medicine. Lastly, by applying tissue engineering and 3D printing techniques, organoids could be used in the future to replace or regenerate damaged tissue. In this review, we will focus on hiPSC-derived 3D cultures and their ability to model human diseases with an in-depth analysis of gene editing applications, as well as tumor models. Furthermore, we will highlight the state-of-the-art of organoid facilities that around the world offer know-how and services. This is an increasing trend that shed the light on the need of bridging the publicand the private sector. Hence, in the context of drug discovery, Organoid Factories can offer biobanks of validated 3D organoid models that can be used in collaboration with pharmaceutical companies to speed up the drug screening process. Finally, we will discuss the limitations and the future development that will lead hiPSC-derived technology from bench to bedside, toward personalized medicine, such as maturity, organoid interconnections, costs, reproducibility and standardization, and ethics. hiPSC-derived organoid technology is now passing from a proof-of-principle to real applications in the clinic, also thanks to the applicability of techniques, such as CRISPR/Cas9 genome editing system, material engineering for the scaffolds, or microfluidic systems. The benefits will have a crucial role in the advance of both basic biological and translational research, particularly in the pharmacological field and drug development. In fact, in the near future, 3D organoids will guide the clinical decision-making process, having validated patient-specific drug screening platforms. This is particularly important in the context of rare genetic diseases or when testing cancer treatments that could in principle have severe side effects. Therefore, this technology has enabled the advancement of personalized medicine in a way never seen before.
Collapse
Affiliation(s)
- Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- IRCCS Neuromed, Pozzilli, IS, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Paola Spitalieri
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Centanini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
17
|
Wang Q, Guo F, Jin Y, Ma Y. Applications of human organoids in the personalized treatment for digestive diseases. Signal Transduct Target Ther 2022; 7:336. [PMID: 36167824 PMCID: PMC9513303 DOI: 10.1038/s41392-022-01194-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/09/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Digestive system diseases arise primarily through the interplay of genetic and environmental influences; there is an urgent need in elucidating the pathogenic mechanisms of these diseases and deploy personalized treatments. Traditional and long-established model systems rarely reproduce either tissue complexity or human physiology faithfully; these shortcomings underscore the need for better models. Organoids represent a promising research model, helping us gain a more profound understanding of the digestive organs; this model can also be used to provide patients with precise and individualized treatment and to build rapid in vitro test models for drug screening or gene/cell therapy, linking basic research with clinical treatment. Over the past few decades, the use of organoids has led to an advanced understanding of the composition of each digestive organ and has facilitated disease modeling, chemotherapy dose prediction, CRISPR-Cas9 genetic intervention, high-throughput drug screening, and identification of SARS-CoV-2 targets, pathogenic infection. However, the existing organoids of the digestive system mainly include the epithelial system. In order to reveal the pathogenic mechanism of digestive diseases, it is necessary to establish a completer and more physiological organoid model. Combining organoids and advanced techniques to test individualized treatments of different formulations is a promising approach that requires further exploration. This review highlights the advancements in the field of organoid technology from the perspectives of disease modeling and personalized therapy.
Collapse
Affiliation(s)
- Qinying Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fanying Guo
- School of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yutao Jin
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Nowak JK, Adams AT, Kalla R, Lindstrøm JC, Vatn S, Bergemalm D, Keita ÅV, Gomollón F, Jahnsen J, Vatn MH, Ricanek P, Ostrowski J, Walkowiak J, Halfvarson J, Satsangi J. Characterisation of the Circulating Transcriptomic Landscape in Inflammatory Bowel Disease Provides Evidence for Dysregulation of Multiple Transcription Factors Including NFE2, SPI1, CEBPB, and IRF2. J Crohns Colitis 2022; 16:1255-1268. [PMID: 35212366 PMCID: PMC9426667 DOI: 10.1093/ecco-jcc/jjac033] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/11/2022] [Accepted: 02/23/2022] [Indexed: 01/11/2023]
Abstract
AIM To assess the pathobiological and translational importance of whole-blood transcriptomic analysis in inflammatory bowel disease [IBD]. METHODS We analysed whole-blood expression profiles from paired-end sequencing in a discovery cohort of 590 Europeans recruited across six countries in the IBD Character initiative (newly diagnosed patients with Crohn's disease [CD; n = 156], ulcerative colitis [UC; n = 167], and controls [n = 267]), exploring differential expression [DESeq2], co-expression networks [WGCNA], and transcription factor involvement [EPEE, ChEA, DoRothEA]. Findings were validated by analysis of an independent replication cohort [99 CD, 100 UC, 95 controls]. In the discovery cohort, we also defined baseline expression correlates of future treatment escalation using cross-validated elastic-net and random forest modelling, along with a pragmatic ratio detection procedure. RESULTS Disease-specific transcriptomes were defined in IBD [8697 transcripts], CD [7152], and UC [8521], with the most highly significant changes in single genes, including CD177 (log2-fold change [LFC] = 4.63, p = 4.05 × 10-118), MCEMP1 [LFC = 2.45, p = 7.37 × 10-109], and S100A12 [LFC = 2.31, p = 2.15 × 10-93]. Significantly over-represented pathways included IL-1 [p = 1.58 × 10-11], IL-4, and IL-13 [p = 8.96 × 10-9]. Highly concordant results were obtained using multiple regulatory activity inference tools applied to the discovery and replication cohorts. These analyses demonstrated central roles in IBD for the transcription factors NFE2, SPI1 [PU.1], CEBPB, and IRF2, all regulators of cytokine signalling, based on a consistent signal across cohorts and transcription factor ranking methods. A number of simple transcriptome-based models were associated with the need for treatment escalation, including the binary CLEC5A/CDH2 expression ratio in UC (hazard ratio = 23.4, 95% confidence interval [CI] 5.3-102.0). CONCLUSIONS Transcriptomic analysis has allowed for a detailed characterisation of IBD pathobiology, with important potential translational implications.
Collapse
Affiliation(s)
- Jan K Nowak
- Corresponding authors: Dr Jan K. Nowak, Translational Gastroenterology Unit, Experimental Medicine Division, John Radcliffe Hospital, Headley Way, Headington, Oxford OX3 9DU, UK.
| | | | - Rahul Kalla
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jonas C Lindstrøm
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Simen Vatn
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Daniel Bergemalm
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Jørgen Jahnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Morten H Vatn
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- EpiGen Institute, Akershus University Hospital, University of Oslo, Oslo, Norway
| | - Petr Ricanek
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Jerzy Ostrowski
- Department of Genetics, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre for Postgraduate Medical Education, Warsaw, Poland
| | - Jaroslaw Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Jack Satsangi
- Jack Satsangi, Translational Gastroenterology Unit, Experimental Medicine Division, John Radcliffe Hospital, Headley Way, Headington, Oxford, OX3 9DU, UK.
| | | |
Collapse
|
19
|
Apical-basal polarity and the control of epithelial form and function. Nat Rev Mol Cell Biol 2022; 23:559-577. [PMID: 35440694 DOI: 10.1038/s41580-022-00465-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 02/02/2023]
Abstract
Epithelial cells are the most common cell type in all animals, forming the sheets and tubes that compose most organs and tissues. Apical-basal polarity is essential for epithelial cell form and function, as it determines the localization of the adhesion molecules that hold the cells together laterally and the occluding junctions that act as barriers to paracellular diffusion. Polarity must also target the secretion of specific cargoes to the apical, lateral or basal membranes and organize the cytoskeleton and internal architecture of the cell. Apical-basal polarity in many cells is established by conserved polarity factors that define the apical (Crumbs, Stardust/PALS1, aPKC, PAR-6 and CDC42), junctional (PAR-3) and lateral (Scribble, DLG, LGL, Yurt and RhoGAP19D) domains, although recent evidence indicates that not all epithelia polarize by the same mechanism. Research has begun to reveal the dynamic interactions between polarity factors and how they contribute to polarity establishment and maintenance. Elucidating these mechanisms is essential to better understand the roles of apical-basal polarity in morphogenesis and how defects in polarity contribute to diseases such as cancer.
Collapse
|
20
|
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
|
21
|
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
|
22
|
Duclaux-Loras R, Lebreton C, Berthelet J, Charbit-Henrion F, Nicolle O, Revenu de Courtils C, Waich S, Valovka T, Khiat A, Rabant M, Racine C, Guerrera IC, Baptista J, Mahe MM, Hess MW, Durel B, Lefort N, Banal C, Parisot M, Talbotec C, Lacaille F, Ecochard-Dugelay E, Demir AM, Vogel GF, Faivre L, Rodrigues A, Fowler D, Janecke AR, Müller T, Huber LA, Rodrigues-Lima F, Ruemmele FM, Uhlig HH, Del Bene F, Michaux G, Cerf-Bensussan N, Parlato M. UNC45A deficiency causes microvillus inclusion disease-like phenotype by impairing myosin VB-dependent apical trafficking. J Clin Invest 2022; 132:154997. [PMID: 35575086 PMCID: PMC9106349 DOI: 10.1172/jci154997] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/29/2022] [Indexed: 01/03/2023] Open
Abstract
Variants in the UNC45A cochaperone have been recently associated with a syndrome combining diarrhea, cholestasis, deafness, and bone fragility. Yet the mechanism underlying intestinal failure in UNC45A deficiency remains unclear. Here, biallelic variants in UNC45A were identified by next-generation sequencing in 6 patients with congenital diarrhea. Corroborating in silico prediction, variants either abolished UNC45A expression or altered protein conformation. Myosin VB was identified by mass spectrometry as client of the UNC45A chaperone and was found misfolded in UNC45AKO Caco-2 cells. In keeping with impaired myosin VB function, UNC45AKO Caco-2 cells showed abnormal epithelial morphogenesis that was restored by full-length UNC45A, but not by mutant alleles. Patients and UNC45AKO 3D organoids displayed altered luminal development and microvillus inclusions, while 2D cultures revealed Rab11 and apical transporter mislocalization as well as sparse and disorganized microvilli. All those features resembled the subcellular abnormalities observed in duodenal biopsies from patients with microvillus inclusion disease. Finally, microvillus inclusions and shortened microvilli were evidenced in enterocytes from unc45a-deficient zebrafish. Taken together, our results provide evidence that UNC45A plays an essential role in epithelial morphogenesis through its cochaperone function of myosin VB and that UNC45A loss causes a variant of microvillus inclusion disease.
Collapse
Affiliation(s)
- Rémi Duclaux-Loras
- Université Paris Cité, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, Paris, France
- Department of Pediatric Gastroenterology, Assistance Publique-Hopitaux de Paris, Hopital Necker–Enfants Malades, F-75015, Paris, France
| | - Corinne Lebreton
- Université Paris Cité, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, Paris, France
| | | | - Fabienne Charbit-Henrion
- Université Paris Cité, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, Paris, France
- Department of Pediatric Gastroenterology, Assistance Publique-Hopitaux de Paris, Hopital Necker–Enfants Malades, F-75015, Paris, France
| | - Ophelie Nicolle
- Université de Rennes, CNRS, Institut de Génétique et Développement de Rennes (IGDR)–UMR 6290, Rennes, France
| | - Céline Revenu de Courtils
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Paris, France
| | - Stephanie Waich
- Universitätsklinik für Pädiatrie I and
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Taras Valovka
- Universitätsklinik für Pädiatrie I and
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Anis Khiat
- Université Paris Cité, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, Paris, France
| | - Marion Rabant
- Department of Pathology, Assistance Publique–Hopitaux de Paris, Hopital Necker–Enfants Malades, Paris, France
| | - Caroline Racine
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Fédération Hospitalo–Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire, and Equipe GAD, Université de Bourgogne Franche-Comté, Faculté de Médecine, INSERM LNC UMR 1231, Dijon, France
| | - Ida Chiara Guerrera
- Proteomics Platform 3P5-Necker, Université Paris Descartes-Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Júlia Baptista
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, United Kingdom
- Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Maxime M. Mahe
- Université de Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Michael W. Hess
- Institut für Histologie und Embryologie Medical University of Innsbruck, Innsbruck, Austria
| | - Béatrice Durel
- Cell Imaging Platform, INSERM-US24-CNRS UMS 3633 Structure Fédérative de Recherche Necker, Université Paris Cité, Paris, France
| | - Nathalie Lefort
- iPS Core Facility, Imagine Institute, INSERM U1163, Paris Descartes University, Paris, France
| | - Céline Banal
- iPS Core Facility, Imagine Institute, INSERM U1163, Paris Descartes University, Paris, France
| | - Mélanie Parisot
- Genomics Core Facility, Institut Imagine–Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Cecile Talbotec
- Department of Pediatric Gastroenterology, Assistance Publique-Hopitaux de Paris, Hopital Necker–Enfants Malades, F-75015, Paris, France
| | - Florence Lacaille
- Department of Pediatric Gastroenterology, Assistance Publique-Hopitaux de Paris, Hopital Necker–Enfants Malades, F-75015, Paris, France
| | | | - Arzu Meltem Demir
- Ankara Child Health and Diseases, Training and Research Hospital, Pediatric Gastroenterology, Ankara, Turkey
| | - Georg F. Vogel
- Universitätsklinik für Pädiatrie I and
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Laurence Faivre
- Department of Pathology, Assistance Publique–Hopitaux de Paris, Hopital Necker–Enfants Malades, Paris, France
| | | | | | | | | | - Lukas A. Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Frank M. Ruemmele
- Department of Pediatric Gastroenterology, Assistance Publique-Hopitaux de Paris, Hopital Necker–Enfants Malades, F-75015, Paris, France
| | - Holm H. Uhlig
- Translational Gastroenterology Unit and Department of Paediatrics, John Radcliffe Hospital, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Paris, France
| | - Grégoire Michaux
- Université de Rennes, CNRS, Institut de Génétique et Développement de Rennes (IGDR)–UMR 6290, Rennes, France
| | - Nadine Cerf-Bensussan
- Université Paris Cité, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, Paris, France
| | - Marianna Parlato
- Université Paris Cité, Imagine Institute, Laboratory of Intestinal Immunity, INSERM, UMR1163, Paris, France
| |
Collapse
|
23
|
Kawasaki M, Goyama T, Tachibana Y, Nagao I, Ambrosini YM. Farm and Companion Animal Organoid Models in Translational Research: A Powerful Tool to Bridge the Gap Between Mice and Humans. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:895379. [PMID: 35647577 PMCID: PMC9133531 DOI: 10.3389/fmedt.2022.895379] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/26/2022] [Indexed: 12/19/2022] Open
Abstract
Animal organoid models derived from farm and companion animals have great potential to contribute to human health as a One Health initiative, which recognize a close inter-relationship among humans, animals and their shared environment and adopt multi-and trans-disciplinary approaches to optimize health outcomes. With recent advances in organoid technology, studies on farm and companion animal organoids have gained more attention in various fields including veterinary medicine, translational medicine and biomedical research. Not only is this because three-dimensional organoids possess unique characteristics from traditional two-dimensional cell cultures including their self-organizing and self-renewing properties and high structural and functional similarities to the originating tissue, but also because relative to conventional genetically modified or artificially induced murine models, companion animal organoids can provide an excellent model for spontaneously occurring diseases which resemble human diseases. These features of companion animal organoids offer a paradigm-shifting approach in biomedical research and improve translatability of in vitro studies to subsequent in vivo studies with spontaneously diseased animals while reducing the use of conventional animal models prior to human clinical trials. Farm animal organoids also could play an important role in investigations of the pathophysiology of zoonotic and reproductive diseases by contributing to public health and improving agricultural production. Here, we discuss a brief history of organoids and the most recent updates on farm and companion animal organoids, followed by discussion on their potential in public health, food security, and comparative medicine as One Health initiatives. We highlight recent evolution in the culturing of organoids and their integration with organ-on-a-chip systems to overcome current limitations in in vitro studies. We envision multidisciplinary work integrating organoid culture and organ-on-a-chip technology can contribute to improving both human and animal health.
Collapse
Affiliation(s)
- Minae Kawasaki
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Takashi Goyama
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Yurika Tachibana
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Itsuma Nagao
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | | |
Collapse
|
24
|
Ramadesikan S, Lee J, Aguilar RC. The Future of Genetic Disease Studies: Assembling an Updated Multidisciplinary Toolbox. Front Cell Dev Biol 2022; 10:886448. [PMID: 35573700 PMCID: PMC9096115 DOI: 10.3389/fcell.2022.886448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
|
25
|
Li Q, Zhou Z, Sun Y, Sun C, Klappe K, van IJzendoorn SC. A Functional Relationship Between UNC45A and MYO5B Connects Two Rare Diseases With Shared Enteropathy. Cell Mol Gastroenterol Hepatol 2022; 14:295-310. [PMID: 35421597 PMCID: PMC9218578 DOI: 10.1016/j.jcmgh.2022.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND & AIMS UNC45A is a myosin (co-)chaperone, and mutations in the UNC45A gene were recently identified in osteo-oto-hepato-enteric (O2HE) syndrome patients presenting with congenital diarrhea and intrahepatic cholestasis. Congenital diarrhea and intrahepatic cholestasis are also the prime symptoms in patients with microvillus inclusion disease (MVID) and mutations in MYO5B, encoding the recycling endosome-associated myosin Vb. The aim of this study was to determine whether UNC45A and myosin Vb are functionally linked. METHODS CRISPR-Cas9 gene editing and site-directed mutagenesis were performed with intestinal epithelial and hepatocellular cell lines, followed by Western blotting, quantitative polymerase chain reaction, and scanning electron and/or confocal fluorescence microscopy to determine the relationship between (mutants of) UNC45A and myosin Vb. RESULTS UNC45A depletion in intestinal and hepatic cells reduced myosin Vb protein expression, and in intestinal epithelial cells, it affected 2 myosin Vb-dependent processes that underlie MVID pathogenesis: rat sarcoma-associated binding protein (RAB)11A-positve recycling endosome positioning and microvilli development. Reintroduction of UNC45A in UNC45A-depleted cells restored myosin Vb expression, and reintroduction of UNC45A or myosin Vb, but not the O2HE patient UNC45A-c.1268T>A variant, restored recycling endosome positioning and microvilli development. The O2HE patient-associated p.V423D substitution, encoded by the UNC45A-c.1268T>A variant, impaired UNC45A protein stability but as such not the ability of UNC45A to promote myosin Vb expression and microvilli development. CONCLUSIONS A functional relationship exists between UNC45A and myosin Vb, thereby connecting 2 rare congenital diseases with overlapping enteropathy at the molecular level. Protein instability rather than functional impairment underlies the pathogenicity of the O2HE syndrome-associated UNC45A-p.V423D mutation.
Collapse
Affiliation(s)
| | | | | | | | | | - Sven C.D. van IJzendoorn
- Correspondence Address correspondence to: Sven C. D. van IJzendoorn, PhD, Department of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
26
|
Nassari S, Lacarrière-Keïta C, Lévesque D, Boisvert FM, Jean S. Rab21 in enterocytes participates in intestinal epithelium maintenance. Mol Biol Cell 2022; 33:ar32. [PMID: 35171715 PMCID: PMC9250356 DOI: 10.1091/mbc.e21-03-0139] [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] [Indexed: 11/11/2022] Open
Abstract
Membrane trafficking is defined as the vesicular transport of proteins into, out of, and throughout the cell. In intestinal enterocytes, defects in endocytic/recycling pathways result in impaired function and are linked to diseases. However, how these trafficking pathways regulate intestinal tissue homeostasis is poorly understood. Using the Drosophila intestine as an in vivo system, we investigated enterocyte-specific functions for the early endosomal machinery. We focused on Rab21, which regulates specific steps in early endosomal trafficking. Depletion of Rab21 in enterocytes led to abnormalities in intestinal morphology, with deregulated cellular equilibrium associated with a gain in mitotic cells and increased cell death. Increases in apoptosis and Yorkie signaling were responsible for compensatory proliferation and tissue inflammation. Using an RNAi screen, we identified regulators of autophagy and membrane trafficking that phenocopied Rab21 knockdown. We further showed that Rab21 knockdown-induced hyperplasia was rescued by inhibition of epidermal growth factor receptor signaling. Moreover, quantitative proteomics identified proteins affected by Rab21 depletion. Of these, we validated changes in apolipoprotein ApoLpp and the trehalose transporter Tret1-1, indicating roles for enterocyte Rab21 in lipid and carbohydrate homeostasis, respectively. Our data shed light on an important role for early endosomal trafficking, and Rab21, in enterocyte-mediated intestinal epithelium maintenance. [Media: see text] [Media: see text].
Collapse
Affiliation(s)
- Sonya Nassari
- Faculté de Médecine et des Sciences de la Santé, Department of Immunology and Cell Biology, Université de Sherbrooke, 3201 Rue Jean Mignault, Sherbrooke, Québec J1E 4K8, Canada
| | - Camille Lacarrière-Keïta
- Faculté de Médecine et des Sciences de la Santé, Department of Immunology and Cell Biology, Université de Sherbrooke, 3201 Rue Jean Mignault, Sherbrooke, Québec J1E 4K8, Canada
| | - Dominique Lévesque
- Faculté de Médecine et des Sciences de la Santé, Department of Immunology and Cell Biology, Université de Sherbrooke, 3201 Rue Jean Mignault, Sherbrooke, Québec J1E 4K8, Canada
| | - François-Michel Boisvert
- Faculté de Médecine et des Sciences de la Santé, Department of Immunology and Cell Biology, Université de Sherbrooke, 3201 Rue Jean Mignault, Sherbrooke, Québec J1E 4K8, Canada
| | - Steve Jean
- Faculté de Médecine et des Sciences de la Santé, Department of Immunology and Cell Biology, Université de Sherbrooke, 3201 Rue Jean Mignault, Sherbrooke, Québec J1E 4K8, Canada
| |
Collapse
|
27
|
Anjum M, Laitila A, Ouwehand AC, Forssten SD. Current Perspectives on Gastrointestinal Models to Assess Probiotic-Pathogen Interactions. Front Microbiol 2022; 13:831455. [PMID: 35173703 PMCID: PMC8841803 DOI: 10.3389/fmicb.2022.831455] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
There are different models available that mimic the human intestinal epithelium and are thus available for studying probiotic and pathogen interactions in the gastrointestinal tract. Although, in vivo models make it possible to study the overall effects of a probiotic on a living subject, they cannot always be conducted and there is a general commitment to reduce the use of animal models. Hence, in vitro methods provide a more rapid tool for studying the interaction between probiotics and pathogens; as well as being ethically superior, faster, and less expensive. The in vitro models are represented by less complex traditional models, standard 2D models compromised of culture plates as well as Transwell inserts, and newer 3D models like organoids, enteroids, as well as organ-on-a-chip. The optimal model selected depends on the research question. Properly designed in vitro and/or in vivo studies are needed to examine the mechanism(s) of action of probiotics on pathogens to obtain physiologically relevant results.
Collapse
Affiliation(s)
| | | | | | - Sofia D. Forssten
- International Flavors and Fragrances, Health and Biosciences, Danisco Sweeteners Oy, Kantvik, Finland
| |
Collapse
|
28
|
Montoya-Cerrillo D, Bernieh A, Saad AG. Critical diagnoses in paediatric gastrointestinal diseases. Pathology 2022; 54:195-206. [PMID: 35033374 DOI: 10.1016/j.pathol.2021.09.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/26/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022]
Abstract
Gastrointestinal biopsies represent an increasing proportion of the paediatric pathologist's workload, an increase fundamentally due to an expansion of the understanding of the basic clinical, molecular, genetic, and histopathological features of paediatric gastrointestinal disorders. The histological interpretation of endoscopically retrieved gastrointestinal biopsies in children requires a unique set of diagnostic expertise and detailed knowledge of various gastrointestinal disorders that have a predilection for the paediatric population. This article's major role is to highlight the unique problems inherent to paediatric gastrointestinal disorders that require immediate communication with the paediatric surgeon or the gastroenterologist. For this, we tried to cover the most important diseases that a paediatric pathologist might encounter in daily practice. Some of these diseases are relatively rare, such as microvillous inclusion disease and tufting enteropathy, but some are more common such as eosinophilic disorders and inflammatory bowel disease. Awareness of the histopathological features of these diseases, particularly those that are relatively uncommon, is crucial to spare the patient a lengthy and costly evaluation. We made a particular effort to abundantly reference this article should the reader wish to expand on the content of any section.
Collapse
Affiliation(s)
| | - Anas Bernieh
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Ali G Saad
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
29
|
Goulet O, Pigneur B, Charbit-Henrion F. Congenital enteropathies involving defects in enterocyte structure or differentiation. Best Pract Res Clin Gastroenterol 2022; 56-57:101784. [PMID: 35331396 DOI: 10.1016/j.bpg.2021.101784] [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: 09/02/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 01/31/2023]
Abstract
Congenital enteropathies (CE) are a group of rare inherited diseases with a typical onset early in life. They involve defects in enterocyte structure or differentiation. They can cause a severe condition of intestinal failure (IF). The diagnostic approach is based first on clinical presentation (consanguinity, prenatal expression, polyhydramnios, early neonatal onset, aspect of stools, persistence at bowel rest, associated extra-digestive manifestations….) and histo-pathological analyses. These rare intestinal diseases cause protracted diarrhea that might resolve, for a few, with a dietetic approach. However, protracted or permanent IF may require long term parenteral nutrition and, in limited cases, intestinal transplantation. With the progresses in both clinical nutrition and genetics, many of these CE are nowadays associated with recognized gene mutations. It improved our knowledge and the understanding in the patho-physiology of these diseases, thus, leading potentially to therapeutic perspectives. These review cover most of the early onset CE and excludes the immune related diarrhea.
Collapse
Affiliation(s)
- Olivier Goulet
- Division of Paediatric Gastroenterology Hepatology and Nutrition, University Paris-Centre, Hôpital Necker-Enfants Malades, 149, Rue de Sèvres, 75743, PARIS Cedex 15, France.
| | - Bénédicte Pigneur
- Division of Paediatric Gastroenterology Hepatology and Nutrition, University Paris-Centre, Hôpital Necker-Enfants Malades, 149, Rue de Sèvres, 75743, PARIS Cedex 15, France
| | - Fabienne Charbit-Henrion
- Department of Genetics, Hôpital Necker-Enfants Malades, 149, Rue de Sèvres, 75743, PARIS Cedex 15, France
| |
Collapse
|
30
|
Griesemer D, Xue JR, Reilly SK, Ulirsch JC, Kukreja K, Davis JR, Kanai M, Yang DK, Butts JC, Guney MH, Luban J, Montgomery SB, Finucane HK, Novina CD, Tewhey R, Sabeti PC. Genome-wide functional screen of 3'UTR variants uncovers causal variants for human disease and evolution. Cell 2021; 184:5247-5260.e19. [PMID: 34534445 PMCID: PMC8487971 DOI: 10.1016/j.cell.2021.08.025] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/25/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022]
Abstract
3' untranslated region (3'UTR) variants are strongly associated with human traits and diseases, yet few have been causally identified. We developed the massively parallel reporter assay for 3'UTRs (MPRAu) to sensitively assay 12,173 3'UTR variants. We applied MPRAu to six human cell lines, focusing on genetic variants associated with genome-wide association studies (GWAS) and human evolutionary adaptation. MPRAu expands our understanding of 3'UTR function, suggesting that simple sequences predominately explain 3'UTR regulatory activity. We adapt MPRAu to uncover diverse molecular mechanisms at base pair resolution, including an adenylate-uridylate (AU)-rich element of LEPR linked to potential metabolic evolutionary adaptations in East Asians. We nominate hundreds of 3'UTR causal variants with genetically fine-mapped phenotype associations. Using endogenous allelic replacements, we characterize one variant that disrupts a miRNA site regulating the viral defense gene TRIM14 and one that alters PILRB abundance, nominating a causal variant underlying transcriptional changes in age-related macular degeneration.
Collapse
Affiliation(s)
- Dustin Griesemer
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA 02115, USA; Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - James R Xue
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02143, USA.
| | - Steven K Reilly
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02143, USA
| | - Jacob C Ulirsch
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kalki Kukreja
- Department of Molecular and Cell Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joe R Davis
- BigHat Biosciences, San Carlos, CA 94070, USA
| | - Masahiro Kanai
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA 02115, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David K Yang
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA
| | - John C Butts
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Mehmet H Guney
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jeremy Luban
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen B Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hilary K Finucane
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carl D Novina
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan Tewhey
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA; Tufts University School of Medicine, Boston, MA 02111, USA
| | - Pardis C Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA 02143, USA; Department Of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02143, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
31
|
Abstract
Organoids are self-organizing, expanding 3D cultures derived from stem cells. Using tissue derived from patients, these miniaturized models recapitulate various aspects of patient physiology and disease phenotypes including genetic profiles and drug sensitivities. As such, patient-derived organoid (PDO) platforms provide an unprecedented opportunity for improving preclinical drug discovery, clinical trial validation, and ultimately patient care. This article reviews the evolution and scope of organoid technology, highlights recent encouraging results using PDOs as potential patient "avatars" to predict drug response and outcomes, and discusses critical parameters for widespread clinical adoption. These include improvements in assay speed, reproducibility, standardization, and automation which are necessary to realize the translational potential of PDOs as clinical tools. The multiple entry points where PDOs may contribute valuable insights in drug discovery and lessen the risks associated with clinical trials are also discussed.
Collapse
Affiliation(s)
- Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27705, USA
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
- Oncode, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina 27705, USA
| |
Collapse
|
32
|
Intestinal immunoregulation: lessons from human mendelian diseases. Mucosal Immunol 2021; 14:1017-1037. [PMID: 33859369 DOI: 10.1038/s41385-021-00398-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.
Collapse
|
33
|
Leng C, Sun Y, van IJzendoorn SCD. Risk and Clinical Significance of Idiopathic Preterm Birth in Microvillus Inclusion Disease. J Clin Med 2021; 10:jcm10173935. [PMID: 34501384 PMCID: PMC8432107 DOI: 10.3390/jcm10173935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Microvillus inclusion disease (MVID) is a rare enteropathy caused by mutations in the MYO5B or STX3 gene. MVID is a disease that is difficult to manage with clinical heterogeneity. Therefore, knowledge about factors influencing MVID morbidity and mortality is urgently needed. Triggered by a recent study that reported a high percentage of preterm births in twelve cases of MVID, we have conducted a comprehensive retrospective study involving 88 cases of MVID with reported gestational ages. We found that moderate to late preterm birth occurred in more than half of all cases, and this was particularly prominent in MYO5B-associated MVID. Preterm birth in MVID counterintuitively correlated with higher birth weight percentiles, and correlated with higher stool outputs and a significantly shorter average survival time. Data from this study thus demonstrate an increased risk of preterm birth in MYO5B-associated MVID, with a clinical impact on morbidity and mortality. Adverse effects associated with preterm birth should be taken into account in the care of children diagnosed with MVID. Documentation of gestational age may contribute to a better prognostic risk assessment in MVID.
Collapse
Affiliation(s)
- Changsen Leng
- Department of Biomedical Sciences of Cells and Systems, Centre for Liver, Digestive and Metabolic Disease, University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (C.L.); (Y.S.)
- Department of Thoracic Surgery, Guangdong Esophageal Cancer Institute, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Centre, Guangzhou 510060, China
| | - Yue Sun
- Department of Biomedical Sciences of Cells and Systems, Centre for Liver, Digestive and Metabolic Disease, University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (C.L.); (Y.S.)
| | - Sven C. D. van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, Centre for Liver, Digestive and Metabolic Disease, University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (C.L.); (Y.S.)
- Correspondence:
| |
Collapse
|
34
|
Pierreux CE. Shaping the thyroid: From peninsula to de novo lumen formation. Mol Cell Endocrinol 2021; 531:111313. [PMID: 33961919 DOI: 10.1016/j.mce.2021.111313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 01/06/2023]
Abstract
A challenging and stimulating question in biology deals with the formation of organs from groups of undifferentiated progenitor cells. Most epithelial organs indeed derive from the endodermal monolayer and evolve into various shape and tridimensional organization adapted to their specialized adult function. Thyroid organogenesis is no exception. In most mammals, it follows a complex and sequential process initiated from the endoderm and leading to the development of a multitude of independent closed spheres equipped and optimized for the synthesis, storage and production of thyroid hormones. The first sign of thyroid organogenesis is visible as a thickening of the anterior foregut endoderm. This group of thyroid progenitors then buds and detaches from the foregut to migrate caudally and then laterally. Upon reaching their final destination in the upper neck region on both sides of the trachea, thyroid progenitors mix with C cell progenitors and finally organize into hormone-producing thyroid follicles. Intrinsic and extrinsic factors controlling thyroid organogenesis have been identified in several species, but the fundamental cellular processes are not sufficiently considered. This review focuses on the cellular aspects of the key morphogenetic steps during thyroid organogenesis and highlights similarities and common mechanisms with developmental steps elucidated in other endoderm-derived organs, despite different final architecture and functions.
Collapse
|
35
|
Genetic Enteropathies Linked to Epithelial Structural Abnormalities and Enteroendocrine Deficiency: A Systematic Review. J Pediatr Gastroenterol Nutr 2021; 72:826-832. [PMID: 33976085 DOI: 10.1097/mpg.0000000000003049] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Congenital diarrhea and enteropathies linked to epithelial structural abnormalities constitute 3 different rare diseases: the tufting enteropathies (TE; EPCAM and SPINT2 mutations), microvillous inclusion disease (MVID; MYO5B and STX3 mutations), and tricho-hepato-enteric syndrome (THE; TTC37 and SKIV2L mutations). Moreover, enteroendocrine deficiencies (ED; PCSK1 and NEUROG3 mutations) share common clinical characteristics with TE, THE, and MVID in that the treatment requires, in most cases, long-term parenteral nutrition. Although numerous cases have been reported in the literature, aggregated data on morbidity and mortality are missing owing to the rarity of the diseases. METHODS We performed a systematic review of all published cases and retrieved 86 articles describing 323 patients (164 boys and 135 girls). RESULTS The mortality rate was 20.28%, with a median age at death of 13.5 months (range 0-228 months); the mortality risk was 30.8/1000 person-year; in half of the cases, death was caused by infections. Parenteral nutrition was required in 95.4% of patients and weaning off from parenteral nutrition was achieved in 29.35% at a median age of 23 months (range 3.3-276 months). The patients with ED linked to PCSK1 were nearly all weaned at a median age of 14 months, but most of the patients became overweight. MVID patients with MYO5B mutations were most often born preterm. ED linked to NEUROG3 mutation and THE patients usually presented with intrauterine growth retardation. CONCLUSIONS This review presents data from 323 patients with congenital diarrhea linked to EPCAM TE, SPINT2 TE, TTC37 THE, SKIV2L THE, MYO5B MVID, STX3 MVID, NEUROG3 ED, and PCSK1 ED mutations.
Collapse
|
36
|
Janecke AR, Liu X, Adam R, Punuru S, Viestenz A, Strauß V, Laass M, Sanchez E, Adachi R, Schatz MP, Saboo US, Mittal N, Rohrschneider K, Escher J, Ganesh A, Al Zuhaibi S, Al Murshedi F, AlSaleem B, Alfadhel M, Al Sinani S, Alkuraya FS, Huber LA, Müller T, Heidelberger R, Janz R. Pathogenic STX3 variants affecting the retinal and intestinal transcripts cause an early-onset severe retinal dystrophy in microvillus inclusion disease subjects. Hum Genet 2021; 140:1143-1156. [PMID: 33974130 PMCID: PMC8263458 DOI: 10.1007/s00439-021-02284-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/15/2021] [Indexed: 01/12/2023]
Abstract
Biallelic STX3 variants were previously reported in five individuals with the severe congenital enteropathy, microvillus inclusion disease (MVID). Here, we provide a significant extension of the phenotypic spectrum caused by STX3 variants. We report ten individuals of diverse geographic origin with biallelic STX3 loss-of-function variants, identified through exome sequencing, single-nucleotide polymorphism array-based homozygosity mapping, and international collaboration. The evaluated individuals all presented with MVID. Eight individuals also displayed early-onset severe retinal dystrophy, i.e., syndromic-intestinal and retinal-disease. These individuals harbored STX3 variants that affected both the retinal and intestinal STX3 transcripts, whereas STX3 variants affected only the intestinal transcript in individuals with solitary MVID. That STX3 is essential for retinal photoreceptor survival was confirmed by the creation of a rod photoreceptor-specific STX3 knockout mouse model which revealed a time-dependent reduction in the number of rod photoreceptors, thinning of the outer nuclear layer, and the eventual loss of both rod and cone photoreceptors. Together, our results provide a link between STX3 loss-of-function variants and a human retinal dystrophy. Depending on the genomic site of a human loss-of-function STX3 variant, it can cause MVID, the novel intestinal-retinal syndrome reported here or, hypothetically, an isolated retinal dystrophy.
Collapse
Affiliation(s)
- Andreas R Janecke
- Department of Pediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
- Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Xiaoqin Liu
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA
| | - Rüdiger Adam
- University Children's Hospital, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Sumanth Punuru
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA
| | - Arne Viestenz
- Department of Ophthalmology, University Medical Center Halle, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Valeria Strauß
- Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Halle, Halle, Germany
| | - Martin Laass
- Klinik und Poliklinik f. Kinder- u. Jugendmedizin, University of Dresden, Dresden, Germany
| | - Elizabeth Sanchez
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto Adachi
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martha P Schatz
- Department of Ophthalmology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ujwala S Saboo
- Department of Ophthalmology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Naveen Mittal
- Department of Department of Pediatrics, Division of Pediatric Gastroenterology, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Johanna Escher
- Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Anuradha Ganesh
- Department of Ophthalmology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Sana Al Zuhaibi
- Department of Ophthalmology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Fathiya Al Murshedi
- Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman
| | - Badr AlSaleem
- King Fahad Medical City, Children's Specialized Hospital, Riyadh, Saudi Arabia
| | - Majid Alfadhel
- Genetics Division and Medical Genomic Research Lab, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Siham Al Sinani
- Department of Child Health, Sultan Qaboos University Hospital, Muscat, Oman
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Lukas A Huber
- Division of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA.
| | - Roger Janz
- Department of Neurobiology and Anatomy, MSB 7.046, McGovern Medical School at the University of Texas HSC (UTHealth), 6431 Fannin Street, Houston, TX, 77030, USA
- Center for Scientific Review, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
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
|
38
|
Kar SK, Wells JM, Ellen ED, Te Pas MFW, Madsen O, Groenen MAM, Woelders H. Organoids: a promising new in vitro platform in livestock and veterinary research. Vet Res 2021; 52:43. [PMID: 33691792 PMCID: PMC7943711 DOI: 10.1186/s13567-021-00904-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Organoids are self-organizing, self-renewing three-dimensional cellular structures that resemble organs in structure and function. They can be derived from adult stem cells, embryonic stem cells, or induced pluripotent stem cells. They contain most of the relevant cell types with a topology and cell-to-cell interactions resembling that of the in vivo tissue. The widespread and increasing adoption of organoid-based technologies in human biomedical research is testament to their enormous potential in basic, translational- and applied-research. In a similar fashion there appear to be ample possibilities for research applications of organoids from livestock and companion animals. Furthermore, organoids as in vitro models offer a great possibility to reduce the use of experimental animals. Here, we provide an overview of studies on organoids in livestock and companion animal species, with focus on the methods developed for organoids from a variety of tissues/organs from various animal species and on the applications in veterinary research. Current limitations, and ongoing research to address these limitations, are discussed. Further, we elaborate on a number of fields of research in animal nutrition, host-microbe interactions, animal breeding and genomics, and animal biotechnology, in which organoids may have great potential as an in vitro research tool.
Collapse
Affiliation(s)
- Soumya K Kar
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands.
| | - Jerry M Wells
- Host-Microbe Interactomics, Wageningen University & Research, Wageningen, The Netherlands
| | - Esther D Ellen
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Marinus F W Te Pas
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, The Netherlands
| | - Martien A M Groenen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, The Netherlands
| | - Henri Woelders
- Wageningen Livestock Research, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
39
|
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: 8] [Impact Index Per Article: 2.0] [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
|
40
|
Fujii M, Sato T. Somatic cell-derived organoids as prototypes of human epithelial tissues and diseases. NATURE MATERIALS 2021; 20:156-169. [PMID: 32807924 DOI: 10.1038/s41563-020-0754-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/30/2020] [Indexed: 05/19/2023]
Abstract
Recent progress in our understanding of the regulation of epithelial tissue stem cells has allowed us to exploit their abilities and instruct them to self-organize into tissue-mimicking structures, so-called organoids. Organoids preserve the molecular, structural and functional characteristics of their tissues of origin, thus providing an attractive opportunity to study the biology of human tissues in health and disease. In parallel to deriving organoids from yet-uncultured epithelial tissues, the field is devoting a growing amount of effort to model human diseases using organoids. This Review describes multidisciplinary approaches for creating organoid models of human genetic, neoplastic, immunological and infectious diseases, and details how they have contributed to our understanding of disease biology. We further highlight the potential role as well as limitations of organoids in clinical practice and showcase the latest achievements and approaches for tuning the organoid culture system to position organoids in biologically defined settings and to grant organoids with better representation of human tissues.
Collapse
Affiliation(s)
- Masayuki Fujii
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Sun L, Rollins D, Qi Y, Fredericks J, Mansell TJ, Jergens A, Phillips GJ, Wannemuehler M, Wang Q. TNFα regulates intestinal organoids from mice with both defined and conventional microbiota. Int J Biol Macromol 2020; 164:548-556. [PMID: 32693143 PMCID: PMC7657954 DOI: 10.1016/j.ijbiomac.2020.07.176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Cytokines are key factors affecting the fate of intestinal stem cells (ISCs) and effective reagents to manipulate ISCs for research purpose. Tumor necrosis factor alpha (TNFα) is a cytokine produced primarily by monocytes and macrophages. It can induce apoptotic cell death and inflammation, and to inhibit tumorigenesis and viral replication. Additionally, TNFα has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). It is therefore important to identify the mechanism by which individual cytokines affect particular cell types. For this purpose, we used both conventional (CONV) and altered Schaedler flora (ASF) C3H/HeN mice to elucidate the effect of different microbial populations (complex versus defined) on growth of miniguts derived from two different intestinal environments. Furthermore, we studied the effects of different concentrations of TNFα extracted from the lymph and spleen on the growth and viability of ISCs recovered from mice bearing the ASF or CONV microbiota. The effect of TNFα on miniguts growth depends not only on the source and concentration, but also on the intestinal microenvironment from which the ISCs were derived. The findings suggest that TNFα influences the proliferation of miniguts derived from ISCs and, therefore, modulates mucosal homeostasis of the host.
Collapse
Affiliation(s)
- Liping Sun
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; School of Environmental & Resource Sciences, Zhejiang A&F University, Lin'an, Zhejiang 311300, China
| | - Derrick Rollins
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; Department of Statistics, Iowa State University, Ames, IA, United States
| | - Yijun Qi
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Jorrell Fredericks
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Thomas J Mansell
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Albert Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Gregory J Phillips
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Michael Wannemuehler
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA, United States
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States.
| |
Collapse
|
44
|
Li Y, Tang P, Cai S, Peng J, Hua G. Organoid based personalized medicine: from bench to bedside. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:21. [PMID: 33135109 PMCID: PMC7603915 DOI: 10.1186/s13619-020-00059-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Three-dimensional cultured organoids have become a powerful in vitro research tool that preserves genetic, phenotypic and behavioral trait of in vivo organs, which can be established from both pluripotent stem cells and adult stem cells. Organoids derived from adult stem cells can be established directly from diseased epithelium and matched normal tissues, and organoids can also be genetically manipulated by CRISPR-Cas9 technology. Applications of organoids in basic research involve the modeling of human development and diseases, including genetic, infectious and malignant diseases. Importantly, accumulating evidence suggests that biobanks of patient-derived organoids for many cancers and cystic fibrosis have great value for drug development and personalized medicine. In addition, organoids hold promise for regenerative medicine. In the present review, we discuss the applications of organoids in the basic and translational research.
Collapse
Affiliation(s)
- Yaqi Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peiyuan Tang
- Institute of Radiation Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Junjie Peng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Guoqiang Hua
- Institute of Radiation Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Cancer institute, Fudan University Shanghai Cancer Center, Shanghai, 230032, China.
| |
Collapse
|
45
|
Campbell JR, Li H, Wang Y, Kozhemyakin M, Hunt AJ, Liu X, Janz R, Heidelberger R. Phosphorylation of the Retinal Ribbon Synapse Specific t-SNARE Protein Syntaxin3B Is Regulated by Light via a Ca 2 +-Dependent Pathway. Front Cell Neurosci 2020; 14:587072. [PMID: 33192329 PMCID: PMC7606922 DOI: 10.3389/fncel.2020.587072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/11/2020] [Indexed: 12/27/2022] Open
Abstract
Neurotransmitter release at retinal ribbon-style synapses utilizes a specialized t-SNARE protein called syntaxin3B (STX3B). In contrast to other syntaxins, STX3 proteins can be phosphorylated in vitro at T14 by Ca2+/calmodulin-dependent protein kinase II (CaMKII). This modification has the potential to modulate SNARE complex formation required for neurotransmitter release in an activity-dependent manner. To determine the extent to which T14 phosphorylation occurs in vivo in the mammalian retina and characterize the pathway responsible for the in vivo phosphorylation of T14, we utilized quantitative immunofluorescence to measure the levels of STX3 and STX3 phosphorylated at T14 (pSTX3) in the synaptic terminals of mouse retinal photoreceptors and rod bipolar cells (RBCs). Results demonstrate that STX3B phosphorylation at T14 is light-regulated and dependent upon the elevation of intraterminal Ca2+. In rod photoreceptor terminals, the ratio of pSTX3 to STX3 was significantly higher in dark-adapted mice, when rods are active, than in light-exposed mice. By contrast, in RBC terminals, the ratio of pSTX3 to STX3 was higher in light-exposed mice, when these terminals are active, than in dark-adapted mice. These results were recapitulated in the isolated eyecup preparation, but only when Ca2+ was included in the external medium. In the absence of external Ca2+, pSTX3 levels remained low regardless of light/dark exposure. Using the isolated RBC preparation, we next showed that elevation of intraterminal Ca2+ alone was sufficient to increase STX3 phosphorylation at T14. Furthermore, both the non-specific kinase inhibitor staurosporine and the selective CaMKII inhibitor AIP inhibited the Ca2+-dependent increase in the pSTX3/STX3 ratio in isolated RBC terminals, while in parallel experiments, AIP suppressed RBC depolarization-evoked exocytosis, measured using membrane capacitance measurements. Our data support a novel, illumination-regulated modulation of retinal ribbon-style synapse function in which activity-dependent Ca2+ entry drives the phosphorylation of STX3B at T14 by CaMKII, which in turn, modulates the ability to form SNARE complexes required for exocytosis.
Collapse
Affiliation(s)
- Joseph R Campbell
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Hongyan Li
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yanzhao Wang
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Maxim Kozhemyakin
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Albert J Hunt
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoqin Liu
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Roger Janz
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
46
|
Russo P. Updates in Pediatric Congenital Enteropathies: Differential Diagnosis, Testing, and Genetics. Surg Pathol Clin 2020; 13:581-600. [PMID: 33183722 DOI: 10.1016/j.path.2020.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Congenital enteropathies comprise a heterogeneous group of disorders typically resulting in severe diarrhea and intestinal failure. Recent advances in and more widespread application of genetic testing have allowed more accurate diagnosis of these entities as well as identification of new disorders, provided a deeper understanding of intestinal pathophysiology through genotype-phenotype correlations, and permitted the exploration of more specific therapies to diseases that have heretofore been resistant to conventional treatments. The therapeutic armamentarium for these disorders now includes intestinal and hematopoietic stem cell transplantation, specific targeted therapy, such as the use of interleukin-1 receptor antagonists and, in some cases, gene therapy. These considerations are particularly applicable to the group of disorders identified as "very-early onset inflammatory bowel disease" (VEO-IBD), for which a veritable explosion of knowledge has occurred in the last decade. The pathologist plays a crucial role in assisting in the diagnosis of these entities and in ruling out other disorders that enter into the differential diagnosis.
Collapse
Affiliation(s)
- Pierre Russo
- Department of Pathology and Laboratory Medicine, Division of Anatomic Pathology, The University of Pennsylvania School of Medicine, The Children's Hospital of Philadelphia, 324 South 34th Street, Main Building, Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
Tang BL. SNAREs and developmental disorders. J Cell Physiol 2020; 236:2482-2504. [PMID: 32959907 DOI: 10.1002/jcp.30067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Members of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family mediate membrane fusion processes associated with vesicular trafficking and autophagy. SNAREs mediate core membrane fusion processes essential for all cells, but some SNAREs serve cell/tissue type-specific exocytic/endocytic functions, and are therefore critical for various aspects of embryonic development. Mutations or variants of their encoding genes could give rise to developmental disorders, such as those affecting the nervous system and immune system in humans. Mutations to components in the canonical synaptic vesicle fusion SNARE complex (VAMP2, STX1A/B, and SNAP25) and a key regulator of SNARE complex formation MUNC18-1, produce variant phenotypes of autism, intellectual disability, movement disorders, and epilepsy. STX11 and MUNC18-2 mutations underlie 2 subtypes of familial hemophagocytic lymphohistiocytosis. STX3 mutations contribute to variant microvillus inclusion disease. Chromosomal microdeletions involving STX16 play a role in pseudohypoparathyroidism type IB associated with abnormal imprinting of the GNAS complex locus. In this short review, I discuss these and other SNARE gene mutations and variants that are known to be associated with a variety developmental disorders, with a focus on their underlying cellular and molecular pathological basis deciphered through disease modeling. Possible pathogenic potentials of other SNAREs whose variants could be disease predisposing are also speculated upon.
Collapse
Affiliation(s)
- Bor L Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
48
|
Badawy A, Elfadul M, Aziabi M, Ageel HI, Aqeel A. Challenges of Microvillus Inclusion Disease in the NICU. Neoreviews 2020; 21:e600-e604. [PMID: 32873653 DOI: 10.1542/neo.21-9-e600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mutations in the myosin 5β, syntaxin-binding protein 2, and syntaxin 3 genes lead to microvillus inclusion disease (MVID), an autosomal recessive congenital enteropathy. This rare disease is characterized by lack of microvilli on the surface of enterocytes in the small intestine, the presence of pathognomonic intracellular microvillus inclusions, and vesicular bodies within these enterocytes. This pathology leads to the characteristic intractable, life-threatening, watery diarrhea. In the more common early-onset form, affected patients present in the first few days after birth, whereas in the late-onset form, clinical manifestations appear at approximately 2 to 3 months of age. Genetic testing can confirm the diagnosis, but the infant's medical history, clinical presentation, and small intestinal biopsy results are strongly suggestive of the diagnosis. The prevalence of MVID is thought to be higher in countries with a high degree of consanguinity. Patients with MVID cannot tolerate feedings and require continuous total parenteral nutrition. Mortality is extremely high in the early-onset type with reports of survival in patients treated with small intestinal transplantation. Medical counseling for parents of infants with MVID needs to reflect our current understanding of the various genetic forms of this disease, the feasible management, and anticipated outcomes.
Collapse
Affiliation(s)
| | | | | | - Hossain Ibrahim Ageel
- Gastroenterology Unit, Pediatrics Department, King Fahd Central Hospital, Gazan, Saudi Arabia
| | | |
Collapse
|
49
|
Pournami F, MK AK, Panackal AV, Nandakumar A, Prabhakar J, Jain N. Microvillus Inclusion Disease: A Rare Mutation of STX3 in Exon 9 Causing Fatal Congenital Diarrheal Disease. J Pediatr Genet 2020; 11:154-157. [DOI: 10.1055/s-0040-1716401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
AbstractInherited diarrheal disorders cause serious morbidity resulting in dependence on intensive care and parenteral nutrition. Microvillus inclusion disease (MVID) has been classically described and results from mutations in the gene coding myosin Vb, which is responsible for enterocyte polarization. Newer reports of mutations resulting in truncated syntaxin 3 (STX3) and Munc18-2 (STXBP2) proteins have been elucidated as causative. To date, five cases of STX3 abnormalities resulting in MVID have been described. We report an infant who presented with congenital diarrhea and was determined to have a rare mutation of STX3. This new finding would be beneficial in future functional genotype–phenotype correlation studies.
Collapse
Affiliation(s)
- Femitha Pournami
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Alok Kumar MK
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Anila V. Panackal
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Anand Nandakumar
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Jyothi Prabhakar
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| | - Naveen Jain
- Department of Neonatology, Kerala Institute of Medical Sciences, Trivandrum, Kerala, India
| |
Collapse
|
50
|
O’Sullivan MJ, Lindsay AJ. The Endosomal Recycling Pathway-At the Crossroads of the Cell. Int J Mol Sci 2020; 21:ijms21176074. [PMID: 32842549 PMCID: PMC7503921 DOI: 10.3390/ijms21176074] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
The endosomal recycling pathway lies at the heart of the membrane trafficking machinery in the cell. It plays a central role in determining the composition of the plasma membrane and is thus critical for normal cellular homeostasis. However, defective endosomal recycling has been linked to a wide range of diseases, including cancer and some of the most common neurological disorders. It is also frequently subverted by many diverse human pathogens in order to successfully infect cells. Despite its importance, endosomal recycling remains relatively understudied in comparison to the endocytic and secretory transport pathways. A greater understanding of the molecular mechanisms that support transport through the endosomal recycling pathway will provide deeper insights into the pathophysiology of disease and will likely identify new approaches for their detection and treatment. This review will provide an overview of the normal physiological role of the endosomal recycling pathway, describe the consequences when it malfunctions, and discuss potential strategies for modulating its activity.
Collapse
|