1
|
Tran SK, Lichtenberg JY, Leonard CE, Williamson JR, Sterling HR, Panek GK, Pearson AH, Lopez S, Lemmon CA, Conway DE, Hwang PY. P-cadherin-dependent adhesions are required for single lumen formation and HGF-mediated cell protrusions during epithelial morphogenesis. iScience 2025; 28:111844. [PMID: 39981519 PMCID: PMC11840494 DOI: 10.1016/j.isci.2025.111844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/09/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
During epithelial morphogenesis, in vivo, epithelial cells form cysts enclosing a single, hollow lumen and extend protrusions as a precursor for tubulogenesis. Cell-cell adhesions (e.g., cadherins) contribute to successful execution of these processes; while there are many different cadherins, one less studied cadherin in epithelial morphogenesis is P-cadherin (CDH3). Here, we investigated the role of CDH3 on successful lumen formation and cell protrusions, using three-dimensional cultures of Madin-Darby canine kidney (MDCK) and CDH3 knockout cell lines. We show that depletion of CDH3 leads to perturbations of hollow lumen formation associated with defects in cell protrusions and tubulogenesis, mediated by Rho/ROCK pathway. CDH3 knockout cells exert lower forces on the surrounding environment compared to wild-type cells, suggesting CDH3 acts as a mechanosensor for stable cell protrusion establishment. Together, our data suggest that CDH3 has an essential function during epithelial morphogenesis by contributing to lumen formation and cell protrusions.
Collapse
Affiliation(s)
- Sydnie K. Tran
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Jessanne Y. Lichtenberg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Corinne E. Leonard
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Jessica R. Williamson
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Hazel R. Sterling
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Grace K. Panek
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA
| | - Amanda H. Pearson
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Santiago Lopez
- Department of Biomedical Engineering, Rice University, Houston, TX 77251, USA
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Daniel E. Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Priscilla Y. Hwang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
2
|
López-García I, Oh S, Chaney C, Tsunezumi J, Drummond I, Oxburgh L, Carroll TJ, Marciano DK. Epithelial tubule interconnection driven by HGF-Met signaling in the kidney. Proc Natl Acad Sci U S A 2024; 121:e2416887121. [PMID: 39705305 PMCID: PMC11670081 DOI: 10.1073/pnas.2416887121] [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: 09/09/2024] [Accepted: 11/11/2024] [Indexed: 12/22/2024] Open
Abstract
The formation of functional epithelial tubules is critical for the development and maintenance of many organ systems. While the mechanisms of tubule formation by epithelial cells are well studied, the process of tubule anastomosis-where tubules connect to form a continuous network-remains poorly understood. In this study, we utilized single-cell RNA sequencing to analyze embryonic mouse kidney tubules undergoing anastomosis. Our analysis identified hepatocyte growth factor (HGF) as a key potential mediator of this process. To investigate this further, we developed an assay using epithelial spheroids with fluorescently tagged apical surfaces, allowing us to visualize and quantify tubule-tubule connections. Our results demonstrate that HGF promotes tubule anastomosis, and it does so through the MAPK signaling pathway and MMPs, independently of cell proliferation. Remarkably, treatment with HGF and collagenase was sufficient to induce tubule anastomosis in embryonic mouse kidneys. These findings provide a foundational understanding of how to enhance the formation of functional tubular networks. This has significant clinical implications for the use of in vitro-grown kidney tissues in transplant medicine, potentially improving the success and integration of transplanted tissues.
Collapse
Affiliation(s)
- Isabel López-García
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75235
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX75235
| | - Sunhee Oh
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75235
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX75235
| | - Christopher Chaney
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX75235
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX7235
| | - Jun Tsunezumi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75235
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX75235
- Molecular Pathology Division, Kanagawa Cancer Center Research Institute, Kanagawa241-8515, Japan
| | - Iain Drummond
- Mount Desert Island Biological Laboratory, Bar Harbor, ME04609
| | | | - Thomas J. Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX7235
| | - Denise K. Marciano
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75235
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX75235
| |
Collapse
|
3
|
Jin X, Lin T, Wang Y, Li X, Yang Y. Functions of p120-catenin in physiology and diseases. Front Mol Biosci 2024; 11:1486576. [PMID: 39498333 PMCID: PMC11532153 DOI: 10.3389/fmolb.2024.1486576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
p120-catenin (p120) plays a vital role in regulating cell-cell adhesion at adherens junctions, interacting with the juxtamembrane domain (JMD) core region of E-cadherin and regulates the stability of cadherin at the cell surface. Previous studies have shown significant functions of p120 in cell-cell adhesion, tumor progression and inflammation. In this review, we will discuss recent progress of p120 in physiological processes and diseases, and focus on the functions of p120 in the regulation of cancer and inflammation.
Collapse
Affiliation(s)
- Xin Jin
- The First Affiliated Hospital (The First School of Clinical Medicine), Guangdong Pharmaceutical University, Guangzhou, China
| | - Ting Lin
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yunjuan Wang
- The First Affiliated Hospital (The First School of Clinical Medicine), Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoqian Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanhong Yang
- The First Affiliated Hospital (The First School of Clinical Medicine), Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
4
|
López-García I, Oh S, Chaney C, Tsunezumi J, Drummond I, Oxburgh L, Carroll T, Marciano DK. Epithelial tubule interconnection driven by HGF-Met signaling in the kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597185. [PMID: 38895378 PMCID: PMC11185679 DOI: 10.1101/2024.06.03.597185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The formation of functional epithelial tubules is a central feature of many organ systems. Although the process of tubule formation by epithelial cells is well-studied, the way in which tubules connect with each other (i.e. anastomose) to form functional networks both in vivo and in vitro is not well understood. A key, unanswered question in the kidney is how the renal vesicles of the embryonic kidney connect with the nascent collecting ducts to form a continuous urinary system. We performed a ligand-receptor pair analysis on single cell RNA-seq data from embryonic mouse kidney tubules undergoing anastomosis to select candidates that might mediate this process in vivo. This analysis identified hepatocyte growth factor (HGF), which has known roles in cell proliferation, migration, and tubulogenesis, as one of several possible candidates. To test this possibility, we designed a novel assay to quantitatively examine epithelial tubule anastomosis in vitro using epithelial spheroids with fluorescently-tagged apical surfaces to enable direct visualization of anastomosis. This revealed that HGF is a potent inducer of tubule anastomosis. Tubule anastomosis occurs through a proliferation-independent mechanism that acts through the MAPK signaling cascade and matrix metalloproteinases (MMPs), the latter suggestive of a role in extracellular matrix turnover. Accordingly, treatment of explanted embryonic mouse kidneys with HGF and collagenase was sufficient to induce kidney tubule anastomosis. These results lay the groundwork for investigating how to promote functional interconnections between tubular epithelia, which have important clinical implications for utilizing in vitro grown kidney tissue in transplant medicine.
Collapse
Affiliation(s)
- Isabel López-García
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Sunhee Oh
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Chris Chaney
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Jun Tsunezumi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Pharmaceutical Sciences, Kyushu University of Health and Welfare, Miyazaki, Japan
| | - Iain Drummond
- Mount Dessert Island Biological Laboratory, Maine, USA
| | - Leif Oxburgh
- Kidney Regenerative Medicine Laboratory, Rogosin Institute, New York, 10021, USA
| | - Thomas Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Denise K. Marciano
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| |
Collapse
|
5
|
Hiremath C, Gao L, Geshow K, Patterson Q, Barlow H, Cleaver O, Marciano DK. Rap1 regulates lumen continuity via Afadin in renal epithelia. Dev Biol 2023; 501:20-27. [PMID: 37276970 PMCID: PMC10460627 DOI: 10.1016/j.ydbio.2023.05.003] [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: 12/02/2021] [Revised: 04/13/2023] [Accepted: 05/11/2023] [Indexed: 06/07/2023]
Abstract
The continuity of a lumen within an epithelial tubule is critical for its function. We previously found that the F-actin binding protein Afadin is required for timely lumen formation and continuity in renal tubules formed from the nephrogenic mesenchyme in mice. Afadin is a known effector and interactor of the small GTPase Rap1, and in the current study, we examine the role of Rap1 in nephron tubulogenesis. Here, we demonstrate that Rap1 is required for nascent lumen formation and continuity in cultured 3D epithelial spheroids and in vivo in murine renal epithelial tubules derived from the nephrogenic mesenchyme, where its absence ultimately leads to severe morphogenetic defects in the tubules. By contrast, Rap1 is not required for lumen continuity or morphogenesis in renal tubules derived from the ureteric epithelium, which differ in that they form by extension from a pre-existing tubule. We further demonstrate that Rap1 is required for correct localization of Afadin to adherens junctions both in vitro and in vivo. Together, these results suggest a model in which Rap1 localizes Afadin to junctional complexes, which in turn regulates nascent lumen formation and positioning to ensure continuous tubulogenesis.
Collapse
Affiliation(s)
- Chitkale Hiremath
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lei Gao
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Kenya Geshow
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Quinten Patterson
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Haley Barlow
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Denise K Marciano
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA.
| |
Collapse
|
6
|
Zhao X, Li X, Sun W, Wei Z, Yu M, Zhang M, Tian R. Case report: "Major fetal cardiac pathology associated with a novel CTNND1 mutation". Front Pediatr 2023; 11:1180381. [PMID: 37274823 PMCID: PMC10235691 DOI: 10.3389/fped.2023.1180381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Background The p120-ctn protein, encoded by CTNND1, is involved in intercellular connections and regulates epithelial-mesenchymal transformation. CTNND1 mutations can lead to blepharocheilodontic syndrome (BCDS). Increasing evidence shows that although BCDS mainly manifests as craniofacial and oral deformities, it can also present as congenital heart disease, limb deformities, and neurodevelopmental disorders. Case description We report a prenatal case of a major cardiac malformation at 24+3 weeks of gestation. Ultrasound examination revealed a hypoplastic left ventricular, aortic coarctation, and a ventricular septal defect. Genetic analysis of the fetal tissues showed the presence of a novel mutation in CTNND1 (NM_001085458.2: c.566_c.567insG; p.Pro190fs*15), which may lead to premature termination of protein coding, while both the parents harbored wild-type CTNND1. To date, only 15 CTNND1 mutations have been reported in 19 patients worldwide, of which approximately 31% (6/19) had a cardiac phenotype. Conclusion To the best of our knowledge, this is the first case report of fetal complicated cardiac malformations caused by this CTNND1 mutation. Our findings provide new clinical references for prenatal diagnosis and suggest an important role for CTNND1 in early cardiac development.
Collapse
Affiliation(s)
- Xuliang Zhao
- Department of Laboratory, The 901th Hospital of the Joint Service of the People's Liberation Army, Hefei, China
| | - Xu Li
- Department of Radiology, Anhui Children’s Hospital, Hefei, China
| | - Weiwei Sun
- Department of Medical, Beijing Chigne Translational Medicine Research Center, Beijing, China
| | - Zhuojun Wei
- Department of Obstetrics and Gynecology, The 901th Hospital of the Joint Service of the People's Liberation Army, Hefei, China
| | - Min Yu
- Department of Obstetrics and Gynecology, The 901th Hospital of the Joint Service of the People's Liberation Army, Hefei, China
| | - Man Zhang
- Department of Laboratory, The 901th Hospital of the Joint Service of the People's Liberation Army, Hefei, China
| | - Ruixia Tian
- Department of Obstetrics and Gynecology, The 901th Hospital of the Joint Service of the People's Liberation Army, Hefei, China
| |
Collapse
|
7
|
Cote LE, Feldman JL. Won't You be My Neighbor: How Epithelial Cells Connect Together to Build Global Tissue Polarity. Front Cell Dev Biol 2022; 10:887107. [PMID: 35800889 PMCID: PMC9253303 DOI: 10.3389/fcell.2022.887107] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial tissues form continuous barriers to protect against external environments. Within these tissues, epithelial cells build environment-facing apical membranes, junction complexes that anchor neighbors together, and basolateral surfaces that face other cells. Critically, to form a continuous apical barrier, neighboring epithelial cells must align their apico-basolateral axes to create global polarity along the entire tissue. Here, we will review mechanisms of global tissue-level polarity establishment, with a focus on how neighboring epithelial cells of different origins align their apical surfaces. Epithelial cells with different developmental origins and/or that polarize at different times and places must align their respective apico-basolateral axes. Connecting different epithelial tissues into continuous sheets or tubes, termed epithelial fusion, has been most extensively studied in cases where neighboring cells initially dock at an apical-to-apical interface. However, epithelial cells can also meet basal-to-basal, posing several challenges for apical continuity. Pre-existing basement membrane between the tissues must be remodeled and/or removed, the cells involved in docking are specialized, and new cell-cell adhesions are formed. Each of these challenges can involve changes to apico-basolateral polarity of epithelial cells. This minireview highlights several in vivo examples of basal docking and how apico-basolateral polarity changes during epithelial fusion. Understanding the specific molecular mechanisms of basal docking is an area ripe for further exploration that will shed light on complex morphogenetic events that sculpt developing organisms and on the cellular mechanisms that can go awry during diseases involving the formation of cysts, fistulas, atresias, and metastases.
Collapse
|
8
|
Yang M, Li S, Huang L, Zhao R, Dai E, Jiang X, He Y, Lu J, Peng L, Liu W, Zhang Z, Jiang D, Zhang Y, Jiang Z, Yang Y, Zhao P, Zhu X, Ding X, Yang Z. CTNND1 variants cause familial exudative vitreoretinopathy through Wnt/Cadherin axis. JCI Insight 2022; 7:158428. [PMID: 35700046 PMCID: PMC9431724 DOI: 10.1172/jci.insight.158428] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a hereditary disorder that can cause vision loss. The CTNND1 gene encodes a cellular adhesion protein p120-catenin (p120), which is essential for vascularization, yet the function of p120 in postnatal physiological angiogenesis remains unclear. Here, we applied whole-exome sequencing (WES) on 140 probands of FEVR families and identified three candidate variants in the human CTNND1 gene. We performed inducible deletion of Ctnnd1 in the postnatal mouse endothelial cells (ECs) and observed typical phenotypes of FEVR. Immunofluorescence of retina flat mounts also revealed immune responses, including reactive astrogliosis and microgliosis accompanied by abnormal Vegfa expression. Using an unbiased proteomics analysis in combination with in vivo or in vitro approaches, we propose that p120 is critical for the integrity of cadherin/catenin complex, and that p120 activates Wnt signaling activity by protecting β-catenin from Gsk3β-ubiquitin-guided degradation. Treatment of CTNND1-depleted HRECs with Gsk3β inhibitors LiCl or CHIR-99021 successfully enhanced cell proliferation by preventing β-catenin from degradation. Moreover, LiCl treatment increased vessel density in Ctnnd1-deficient mouse retinas. Functional analysis also revealed that variants in CTNND1 cause FEVR by compromising the expression of adherens junctions (AJs) and Wnt signaling activity. Additionally, genetic interactions between p120 and β-catenin or α-catenin revealed by double heterozygous deletion in mice further confirmed that p120 regulates vascular development through the Wnt/Cadherin axis. Together, we propose that CTNND1 is a novel candidate gene associated with FEVR, and that variants in CTNND1 can cause FEVR through the Wnt/Cadherin axis.
Collapse
Affiliation(s)
- Mu Yang
- Prenatal Diagnosis Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujin Li
- Prenatal Diagnosis Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rulian Zhao
- Prenatal Diagnosis Center, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Erkuan Dai
- Department of Ophthalmology, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Chengdu, China
| | - Xiaoyan Jiang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunqi He
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinglin Lu
- Prenatal Diagnosis Center, Sun Yat-sen University, Guangzhou, China
| | - Li Peng
- Center for Human Molecular Genetics, Sun Yat-sen University, Chengdu, China
| | - Wenjing Liu
- Center for Human Molecular Genetics, Sun Yat-sen University, Chengdu, China
| | - Zhaotian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Dan Jiang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Sichua, Chengdu, China
| | - Yi Zhang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhilin Jiang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Yeming Yang
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Peiquan Zhao
- Department of Ophthalmology, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Chengdu, China
| | - Xianjun Zhu
- Center for Human Molecular Genetics, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhenglin Yang
- Department of Medical Genetics, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
9
|
Alharatani R, Ververi A, Beleza-Meireles A, Ji W, Mis E, Patterson QT, Griffin JN, Bhujel N, Chang CA, Dixit A, Konstantino M, Healy C, Hannan S, Neo N, Cash A, Li D, Bhoj E, Zackai EH, Cleaver R, Baralle D, McEntagart M, Newbury-Ecob R, Scott R, Hurst JA, Au PYB, Hosey MT, Khokha M, Marciano DK, Lakhani SA, Liu KJ. Novel truncating mutations in CTNND1 cause a dominant craniofacial and cardiac syndrome. Hum Mol Genet 2021; 29:1900-1921. [PMID: 32196547 PMCID: PMC7372553 DOI: 10.1093/hmg/ddaa050] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/18/2022] Open
Abstract
CTNND1 encodes the p120-catenin (p120) protein, which has a wide range of functions, including the maintenance of cell–cell junctions, regulation of the epithelial-mesenchymal transition and transcriptional signalling. Due to advances in next-generation sequencing, CTNND1 has been implicated in human diseases including cleft palate and blepharocheilodontic (BCD) syndrome albeit only recently. In this study, we identify eight novel protein-truncating variants, six de novo, in 13 participants from nine families presenting with craniofacial dysmorphisms including cleft palate and hypodontia, as well as congenital cardiac anomalies, limb dysmorphologies and neurodevelopmental disorders. Using conditional deletions in mice as well as CRISPR/Cas9 approaches to target CTNND1 in Xenopus, we identified a subset of phenotypes that can be linked to p120-catenin in epithelial integrity and turnover, and additional phenotypes that suggest mesenchymal roles of CTNND1. We propose that CTNND1 variants have a wider developmental role than previously described and that variations in this gene underlie not only cleft palate and BCD but may be expanded to a broader velocardiofacial-like syndrome.
Collapse
Affiliation(s)
- Reham Alharatani
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK.,Paediatric Dentistry, Centre of Oral, Clinical and Translational Science, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE5 9RS, UK
| | - Athina Ververi
- Department of Clinical Genetics, Great Ormond Street Hospital Trust, London WC1N 3JH, UK
| | - Ana Beleza-Meireles
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK.,Department of Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London SE1 9RT, UK
| | - Weizhen Ji
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Emily Mis
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Quinten T Patterson
- Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8856, USA
| | - John N Griffin
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK.,Pediatric Genomics Discovery Program, Departments of Genetics and Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Nabina Bhujel
- South Thames Cleft Service, Guy's and St. Thomas' NHS Foundation Trust, London SE1 7EH, UK
| | - Caitlin A Chang
- Department of Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, AB, Canada
| | - Abhijit Dixit
- Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB, UK
| | - Monica Konstantino
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Christopher Healy
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Sumayyah Hannan
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| | - Natsuko Neo
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK.,Tokyo Medical and Dental University, Tokyo, Japan
| | - Alex Cash
- South Thames Cleft Service, Guy's and St. Thomas' NHS Foundation Trust, London SE1 7EH, UK
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elizabeth Bhoj
- Department of Pediatrics, Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elaine H Zackai
- Department of Pediatrics, Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ruth Cleaver
- Peninsula Clinical Genetics Service, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK
| | - Diana Baralle
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Meriel McEntagart
- Department of Clinical Genetics, St George's Hospital, London SW17 0RE, UK
| | - Ruth Newbury-Ecob
- Clinical Genetics, University Hospital Bristol NHS Foundation Trust, Bristol BS2 8EG, UK
| | - Richard Scott
- Department of Clinical Genetics, Great Ormond Street Hospital Trust, London WC1N 3JH, UK
| | - Jane A Hurst
- Department of Clinical Genetics, Great Ormond Street Hospital Trust, London WC1N 3JH, UK
| | - Ping Yee Billie Au
- Department of Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, AB, Canada
| | - Marie Therese Hosey
- Paediatric Dentistry, Centre of Oral, Clinical and Translational Science, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE5 9RS, UK
| | - Mustafa Khokha
- Pediatric Genomics Discovery Program, Departments of Genetics and Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Denise K Marciano
- Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8856, USA
| | - Saquib A Lakhani
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE1 9RT, UK
| |
Collapse
|
10
|
Krneta-Stankic V, Corkins ME, Paulucci-Holthauzen A, Kloc M, Gladden AB, Miller RK. The Wnt/PCP formin Daam1 drives cell-cell adhesion during nephron development. Cell Rep 2021; 36:109340. [PMID: 34233186 PMCID: PMC8629027 DOI: 10.1016/j.celrep.2021.109340] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/31/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
E-cadherin junctions facilitate assembly and disassembly of cell contacts that drive development and homeostasis of epithelial tissues. In this study, using Xenopus embryonic kidney and Madin-Darby canine kidney (MDCK) cells, we investigate the role of the Wnt/planar cell polarity (PCP) formin Daam1 (Dishevelled-associated activator of morphogenesis 1) in regulating E-cadherin-based intercellular adhesion. Using live imaging, we show that Daam1 localizes to newly formed cell contacts in the developing nephron. Furthermore, analyses of junctional filamentous actin (F-actin) upon Daam1 depletion indicate decreased microfilament localization and slowed turnover. We also show that Daam1 is necessary for efficient and timely localization of junctional E-cadherin, mediated by Daam1’s formin homology domain 2 (FH2). Finally, we establish that Daam1 signaling promotes organized movement of renal cells. This study demonstrates that Daam1 formin junctional activity is critical for epithelial tissue organization. How cells remodel their adhesions through cell-surface proteins such as E-cadherin is a central question in epithelial tissue biology. Krneta-Stankic et al. show that the Wnt/PCP formin Daam1 regulates cytoskeletal membrane dynamics and E-cadherin localization within developing nephrons. These findings provide a new framework for studying cell-cell adhesion and nephron morphogenesis.
Collapse
Affiliation(s)
- Vanja Krneta-Stankic
- Program in Genes and Development, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Mark E Corkins
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | | | - Malgorzata Kloc
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - Andrew B Gladden
- Program in Genes and Development, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rachel K Miller
- Program in Genes and Development, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston, TX 77030, USA; Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Program in Biochemistry and Cell Biology, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Li H, Hohenstein P, Kuure S. Embryonic Kidney Development, Stem Cells and the Origin of Wilms Tumor. Genes (Basel) 2021; 12:genes12020318. [PMID: 33672414 PMCID: PMC7926385 DOI: 10.3390/genes12020318] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
The adult mammalian kidney is a poorly regenerating organ that lacks the stem cells that could replenish functional homeostasis similarly to, e.g., skin or the hematopoietic system. Unlike a mature kidney, the embryonic kidney hosts at least three types of lineage-specific stem cells that give rise to (a) a ureter and collecting duct system, (b) nephrons, and (c) mesangial cells together with connective tissue of the stroma. Extensive interest has been raised towards these embryonic progenitor cells, which are normally lost before birth in humans but remain part of the undifferentiated nephrogenic rests in the pediatric renal cancer Wilms tumor. Here, we discuss the current understanding of kidney-specific embryonic progenitor regulation in the innate environment of the developing kidney and the types of disruptions in their balanced regulation that lead to the formation of Wilms tumor.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Peter Hohenstein
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
- GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-2941-59395
| |
Collapse
|
12
|
Grimsley-Myers CM, Isaacson RH, Cadwell CM, Campos J, Hernandes MS, Myers KR, Seo T, Giang W, Griendling KK, Kowalczyk AP. VE-cadherin endocytosis controls vascular integrity and patterning during development. J Cell Biol 2021; 219:151601. [PMID: 32232465 PMCID: PMC7199849 DOI: 10.1083/jcb.201909081] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/10/2020] [Accepted: 02/28/2020] [Indexed: 12/18/2022] Open
Abstract
Tissue morphogenesis requires dynamic intercellular contacts that are subsequently stabilized as tissues mature. The mechanisms governing these competing adhesive properties are not fully understood. Using gain- and loss-of-function approaches, we tested the role of p120-catenin (p120) and VE-cadherin (VE-cad) endocytosis in vascular development using mouse mutants that exhibit increased (VE-cadGGG/GGG) or decreased (VE-cadDEE/DEE) internalization. VE-cadGGG/GGG mutant mice exhibited reduced VE-cad-p120 binding, reduced VE-cad levels, microvascular hemorrhaging, and decreased survival. By contrast, VE-cadDEE/DEE mutants exhibited normal vascular permeability but displayed microvascular patterning defects. Interestingly, VE-cadDEE/DEE mutant mice did not require endothelial p120, demonstrating that p120 is dispensable in the context of a stabilized cadherin. In vitro, VE-cadDEE mutant cells displayed defects in polarization and cell migration that were rescued by uncoupling VE-cadDEE from actin. These results indicate that cadherin endocytosis coordinates cell polarity and migration cues through actin remodeling. Collectively, our results indicate that regulated cadherin endocytosis is essential for both dynamic cell movements and establishment of stable tissue architecture.
Collapse
Affiliation(s)
| | - Robin H Isaacson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Chantel M Cadwell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Jazmin Campos
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Marina S Hernandes
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Kenneth R Myers
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Tadahiko Seo
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - William Giang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Andrew P Kowalczyk
- Department of Cell Biology, Department of Dermatology, and Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
13
|
Narayanan V, Schappell LE, Mayer CR, Duke AA, Armiger TJ, Arsenovic PT, Mohan A, Dahl KN, Gleghorn JP, Conway DE. Osmotic Gradients in Epithelial Acini Increase Mechanical Tension across E-cadherin, Drive Morphogenesis, and Maintain Homeostasis. Curr Biol 2020; 30:624-633.e4. [PMID: 31983640 DOI: 10.1016/j.cub.2019.12.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 10/04/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022]
Abstract
Epithelial cells spontaneously form acini (also known as cysts or spheroids) with a single, fluid-filled central lumen when grown in 3D matrices. The size of the lumen is dependent on apical secretion of chloride ions, most notably by the CFTR channel, which has been suggested to establish pressure in the lumen due to water influx. To study the cellular biomechanics of acini morphogenesis and homeostasis, we used MDCK-2 cells. Using FRET-force biosensors for E-cadherin, we observed significant increases in the average tension per molecule for each protein in mature 3D acini as compared to 2D monolayers. Increases in CFTR activity resulted in increased E-cadherin forces, indicating that ionic gradients affect cellular tension. Direct measurements of pressure revealed that mature acini experience significant internal hydrostatic pressure (37 ± 10.9 Pa). Changes in CFTR activity resulted in pressure and/or volume changes, both of which affect E-cadherin tension. Increases in CFTR chloride secretion also induced YAP signaling and cellular proliferation. In order to recapitulate disruption of acinar homeostasis, we induced epithelial-to-mesenchymal transition (EMT). During the initial stages of EMT, there was a gradual decrease in E-cadherin force and lumen pressure that correlated with lumen infilling. Strikingly, increasing CFTR activity was sufficient to block EMT. Our results show that ion secretion is an important regulator of morphogenesis and homeostasis in epithelial acini. Furthermore, this work demonstrates that, for closed 3D cellular systems, ion gradients can generate osmotic pressure or volume changes, both of which result in increased cellular tension.
Collapse
Affiliation(s)
- Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Laurel E Schappell
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Carl R Mayer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Ashley A Duke
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Travis J Armiger
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Paul T Arsenovic
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Abhinav Mohan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Kris N Dahl
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA.
| |
Collapse
|
14
|
Kuure S, Sariola H. Mouse Models of Congenital Kidney Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:109-136. [PMID: 32304071 DOI: 10.1007/978-981-15-2389-2_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects, which cause the majority of chronic kidney diseases in children. CAKUT covers a wide range of malformations that derive from deficiencies in embryonic kidney and lower urinary tract development, including renal aplasia, hypodysplasia, hypoplasia, ectopia, and different forms of ureter abnormalities. The majority of the genetic causes of CAKUT remain unknown. Research on mutant mice has identified multiple genes that critically regulate renal differentiation. The data generated from this research have served as an excellent resource to identify the genetic bases of human kidney defects and have led to significantly improved diagnostics. Furthermore, genetic data from human CAKUT studies have also revealed novel genes regulating kidney differentiation.
Collapse
Affiliation(s)
- Satu Kuure
- GM-Unit, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland. .,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Hannu Sariola
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Paediatric Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
15
|
Abstract
Epithelial-to-mesenchymal transitions (EMTs) require a complete reorganization of cadherin-based cell-cell junctions. p120-catenin binds to the cytoplasmic juxtamembrane domain of classical cadherins and regulates their stability, suggesting that p120-catenin may play an important role in EMTs. Here, we describe the role of p120-catenin in mouse gastrulation, an EMT that can be imaged at cellular resolution and is accessible to genetic manipulation. Mouse embryos that lack all p120-catenin, or that lack p120-catenin in the embryo proper, survive to midgestation. However, mutants have specific defects in gastrulation, including a high rate of p53-dependent cell death, a bifurcation of the posterior axis, and defects in the migration of mesoderm; all are associated with abnormalities in the primitive streak, the site of the EMT. In embryonic day 7.5 (E7.5) mutants, the domain of expression of the streak marker Brachyury (T) expands more than 3-fold, from a narrow strip of posterior cells to encompass more than one-quarter of the embryo. After E7.5, the enlarged T+ domain splits in 2, separated by a mass of mesoderm cells. Brachyury is a direct target of canonical WNT signaling, and the domain of WNT response in p120-catenin mutant embryos, like the T domain, is first expanded, and then split, and high levels of nuclear β-catenin levels are present in the cells of the posterior embryo that are exposed to high levels of WNT ligand. The data suggest that p120-catenin stabilizes the membrane association of β-catenin, thereby preventing accumulation of nuclear β-catenin and excessive activation of the WNT pathway during EMT.
Collapse
|
16
|
Motahari Z, Moody SA, Maynard TM, LaMantia AS. In the line-up: deleted genes associated with DiGeorge/22q11.2 deletion syndrome: are they all suspects? J Neurodev Disord 2019; 11:7. [PMID: 31174463 PMCID: PMC6554986 DOI: 10.1186/s11689-019-9267-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 04/21/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND 22q11.2 deletion syndrome (22q11DS), a copy number variation (CNV) disorder, occurs in approximately 1:4000 live births due to a heterozygous microdeletion at position 11.2 (proximal) on the q arm of human chromosome 22 (hChr22) (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011). This disorder was known as DiGeorge syndrome, Velo-cardio-facial syndrome (VCFS) or conotruncal anomaly face syndrome (CTAF) based upon diagnostic cardiovascular, pharyngeal, and craniofacial anomalies (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011; Burn et al., J Med Genet 30:822-4, 1993) before this phenotypic spectrum was associated with 22q11.2 CNVs. Subsequently, 22q11.2 deletion emerged as a major genomic lesion associated with vulnerability for several clinically defined behavioral deficits common to a number of neurodevelopmental disorders (Fernandez et al., Principles of Developmental Genetics, 2015; Robin and Shprintzen, J Pediatr 147:90-6, 2005; Schneider et al., Am J Psychiatry 171:627-39, 2014). RESULTS The mechanistic relationships between heterozygously deleted 22q11.2 genes and 22q11DS phenotypes are still unknown. We assembled a comprehensive "line-up" of the 36 protein coding loci in the 1.5 Mb minimal critical deleted region on hChr22q11.2, plus 20 protein coding loci in the distal 1.5 Mb that defines the 3 Mb typical 22q11DS deletion. We categorized candidates based upon apparent primary cell biological functions. We analyzed 41 of these genes that encode known proteins to determine whether haploinsufficiency of any single 22q11.2 gene-a one gene to one phenotype correspondence due to heterozygous deletion restricted to that locus-versus complex multigenic interactions can account for single or multiple 22q11DS phenotypes. CONCLUSIONS Our 22q11.2 functional genomic assessment does not support current theories of single gene haploinsufficiency for one or all 22q11DS phenotypes. Shared molecular functions, convergence on fundamental cell biological processes, and related consequences of individual 22q11.2 genes point to a matrix of multigenic interactions due to diminished 22q11.2 gene dosage. These interactions target fundamental cellular mechanisms essential for development, maturation, or homeostasis at subsets of 22q11DS phenotypic sites.
Collapse
Affiliation(s)
- Zahra Motahari
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Sally Ann Moody
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Thomas Michael Maynard
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| | - Anthony-Samuel LaMantia
- The Institute for Neuroscience, and Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 USA
| |
Collapse
|
17
|
Differential expression of p120-catenin 1 and 3 isoforms in epithelial tissues. Sci Rep 2019; 9:90. [PMID: 30643202 PMCID: PMC6331582 DOI: 10.1038/s41598-018-36889-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/28/2018] [Indexed: 01/21/2023] Open
Abstract
P120 catenin (p120) is a non-redundant master regulatory protein of cadherin-based cell-cell junctions, intracellular signaling, and tissue homeostasis and repair. Alternative splicing can generate p120 isoforms 1 and 3 (p120-1 and p120-3), which are implicated in non-overlapping functions by differential expression regulation and unique interactions in different cell types, with often predominant expression of p120-1 in mesenchymal cells, and p120-3 generally prevalent in epithelial cells. However, the lack of specific p120-3 protein detection has precluded analysis of their relative abundance in tissues. Here, we have developed a p120-3 isoform-specific antibody and analyzed the p120-3 localization relative to p120-1 in human tissues. p120-3 but not p120-1 is highly expressed in cell-cell junctions of simple gastrointestinal epithelia such as colon and stomach, and the acini of salivary glands and the pancreas. Conversely, the basal layer of the epidermis and hair follicles expressed p120-1 with reduced p120-3, whereas most other epithelia co-expressed p120-3 and p120-1, including bronchial epithelia and mammary luminal epithelial cells. These data provide an inventory of tissue-specific p120 isoform expression and suggest a link between p120 isoform expression and epithelial differentiation.
Collapse
|
18
|
Establishment of renal proximal tubule cell lines derived from the kidney of p53 knockout mice. Cytotechnology 2019; 71:45-56. [PMID: 30603921 DOI: 10.1007/s10616-018-0261-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
The human cell line HK-2 is most commonly used as a model of renal proximal tubular epithelial cells (PTECs) for various studies despite the absence or low expression of transporters characteristic of parental PTECs. In an effort to develop reliable PTEC models, several human cell lines have been newly established over the last decade. In contrast, reliable mouse PTEC models are still unavailable. In this study, we established immortalized renal cortex tubule cell lines derived from p53 knockout mice and evaluated various PTEC characteristics toward the development of reliable mouse PTEC models. Here, we focus on MuRTE61, one of 13 newly established clonal cell lines. Albumin uptake in MuRTE61 cells was verified by incubation with fluorescent dye-labeled albumin. RT-PCR confirmed the expression of efflux transporter genes characteristic of PTECs in the MuRTE61 cells. MuRTE61 cells exhibited high sensitivity to treatment with cisplatin, a nephrotoxic agent, accompanied by upregulated expression of the uptake transporter Slc22a2 gene. Furthermore, MuRTE61 cells consistently formed spheroids with a lumen and apicobasal polarity in three-dimensional Matrigel cultures. Apical brush border microvilli were also observed in the spheroids by transmission electron microscopy. These data validate that MuRTE61 can be characterized as a reliable mouse PTEC line. In future, detailed analysis of reliable mouse and human PTEC lines will provide an accurate extrapolation of results of experiments using mice and humans, and may help resolve apparent inconsistencies between mouse and human nephrotoxicity.
Collapse
|
19
|
Puri P, Schaefer CM, Bushnell D, Taglienti ME, Kreidberg JA, Yoder BK, Bates CM. Ectopic Phosphorylated Creb Marks Dedifferentiated Proximal Tubules in Cystic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:84-94. [PMID: 29107072 PMCID: PMC5745541 DOI: 10.1016/j.ajpath.2017.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/18/2017] [Accepted: 09/11/2017] [Indexed: 01/25/2023]
Abstract
Ectopic cAMP signaling is pathologic in polycystic kidney disease; however, its spatiotemporal actions are unclear. We characterized the expression of phosphorylated Creb (p-Creb), a target and mediator of cAMP signaling, in developing and cystic kidney models. We also examined tubule-specific effects of cAMP analogs in cystogenesis in embryonic kidney explants. In wild-type mice, p-Creb marked nephron progenitors (NP), early epithelial NP derivatives, ureteric bud, and cortical stroma; p-Creb was present in differentiated thick ascending limb of Henle, collecting duct, and stroma; however, it disappeared in mature NP-derived proximal tubules. In Six2cre;Frs2αFl/Fl mice, a renal cystic model, ectopic p-Creb stained proximal tubule-derived cystic segments that lost the differentiation marker lotus tetragonolobus lectin. Furthermore, lotus tetragonolobus lectin-negative/p-Creb-positive cyst segments (re)-expressed Ncam1, Pax2, and Sox9 markers of immature nephron structures and dedifferentiated proximal tubules after acute kidney injury. These dedifferentiation markers were co-expressed with p-Creb in renal cysts in Itf88 knockout mice subjected to ischemia and Six2cre;Pkd1Fl/Fl mice, other renal cystogenesis models. 8-Br-cAMP addition to wild-type embryonic kidney explants induced proximal tubular cystogenesis and p-Creb expression; these effects were blocked by co-addition of protein kinase A inhibitor. Thus p-Creb/cAMP signaling is appropriate in NP and early nephron derivatives, but disappears in mature proximal tubules. Moreover, ectopic p-Creb expression/cAMP signaling marks dedifferentiated proximal tubular cystic segments. Furthermore, proximal tubules are predisposed to become cystic after cAMP stimulation.
Collapse
Affiliation(s)
- Pawan Puri
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Caitlin M Schaefer
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel Bushnell
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mary E Taglienti
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts; Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Jordan A Kreidberg
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts; Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Bradley K Yoder
- Department of Pediatrics, Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carlton M Bates
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Division of Nephrology, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
20
|
Gao L, Yang Z, Hiremath C, Zimmerman SE, Long B, Brakeman PR, Mostov KE, Bryant DM, Luby-Phelps K, Marciano DK. Afadin orients cell division to position the tubule lumen in developing renal tubules. Development 2017; 144:3511-3520. [PMID: 28860115 DOI: 10.1242/dev.148908] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 08/11/2017] [Indexed: 01/05/2023]
Abstract
In many types of tubules, continuity of the lumen is paramount to tubular function, yet how tubules generate lumen continuity in vivo is not known. We recently found that the F-actin-binding protein afadin is required for lumen continuity in developing renal tubules, though its mechanism of action remains unknown. Here, we demonstrate that afadin is required for lumen continuity by orienting the mitotic spindle during cell division. Using an in vitro 3D cyst model, we find that afadin localizes to the cell cortex adjacent to the spindle poles and orients the mitotic spindle. In tubules, cell division may be oriented relative to two axes: longitudinal and apical-basal. Unexpectedly, in vivo examination of early-stage developing nephron tubules reveals that cell division is not oriented in the longitudinal (or planar-polarized) axis. However, cell division is oriented perpendicular to the apical-basal axis. Absence of afadin in vivo leads to misorientation of apical-basal cell division in nephron tubules. Together, these results support a model whereby afadin determines lumen placement by directing apical-basal spindle orientation, resulting in a continuous lumen and normal tubule morphogenesis.
Collapse
Affiliation(s)
- Lei Gao
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhufeng Yang
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chitkale Hiremath
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Susan E Zimmerman
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Blake Long
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Paul R Brakeman
- Department of Pediatrics, University of California, San Francisco, CA, 94158, USA
| | - Keith E Mostov
- Department of Anatomy, University of California, San Francisco, CA, 94158, USA
| | - David M Bryant
- Institute of Cancer Sciences, University of Glasgow and Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | | | - Denise K Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
21
|
Balasubramaniam SL, Gopalakrishnapillai A, Petrelli NJ, Barwe SP. Knockdown of sodium-calcium exchanger 1 induces epithelial-to-mesenchymal transition in kidney epithelial cells. J Biol Chem 2017; 292:11388-11399. [PMID: 28550085 DOI: 10.1074/jbc.m116.752352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 05/17/2017] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal-to-epithelial transition (MET) and epithelial-to-mesenchymal transition (EMT) are important processes in kidney development. Failure to undergo MET during development leads to the initiation of Wilms tumor, whereas EMT contributes to the development of renal cell carcinomas (RCC). The role of calcium regulators in governing these processes is becoming evident. We demonstrated earlier that Na+/Ca2+ exchanger 1 (NCX1), a major calcium exporter in renal epithelial cells, regulates epithelial cell motility. Here, we show for the first time that NCX1 mRNA and protein expression was down-regulated in Wilms tumor and RCC. Knockdown of NCX1 in Madin-Darby canine kidney cells induced fibroblastic morphology, increased intercellular junctional distance, and induced paracellular permeability, loss of apico-basal polarity in 3D cultures, and anchorage-independent growth, accompanied by expression of mesenchymal markers. We also provide evidence that NCX1 interacts with and anchors E-cadherin to the cell surface independent of NCX1 ion transport activity. Consistent with destabilization of E-cadherin, NCX1 knockdown cells showed an increase in β-catenin nuclear localization, enhanced transcriptional activity, and up-regulation of downstream targets of the β-catenin signaling pathway. Taken together, knockdown of NCX1 in Madin-Darby canine kidney cells alters epithelial morphology and characteristics by destabilization of E-cadherin and induction of β-catenin signaling.
Collapse
Affiliation(s)
- Sona Lakshme Balasubramaniam
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803.,the Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, and
| | - Anilkumar Gopalakrishnapillai
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803
| | - Nicholas J Petrelli
- the Helen F. Graham Cancer Center, Christiana Care Health System, Newark, Delaware 19718
| | - Sonali P Barwe
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803, .,the Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, and
| |
Collapse
|
22
|
Duñach M, Del Valle-Pérez B, García de Herreros A. p120-catenin in canonical Wnt signaling. Crit Rev Biochem Mol Biol 2017; 52:327-339. [PMID: 28276699 DOI: 10.1080/10409238.2017.1295920] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Canonical Wnt signaling controls β-catenin protein stabilization, its translocation to the nucleus and the activation of β-catenin/Tcf-4-dependent transcription. In this review, we revise and discuss the recent results describing actions of p120-catenin in different phases of this pathway. More specifically, we comment its involvement in four different steps: (i) the very early activation of CK1ɛ, essential for Dvl-2 binding to the Wnt receptor complex; (ii) the internalization of GSK3 and Axin into multivesicular bodies, necessary for a complete stabilization of β-catenin; (iii) the activation of Rac1 small GTPase, required for β-catenin translocation to the nucleus; and (iv) the release of the inhibitory action caused by Kaiso transcriptional repressor. We integrate these new results with the previously known action of other elements in this pathway, giving a particular relevance to the responses of the Wnt pathway not required for β-catenin stabilization but for β-catenin transcriptional activity. Moreover, we discuss the possible future implications, suggesting that the two cellular compartments where β-catenin is localized, thus, the adherens junction complex and the Wnt signalosome, are more physically connected that previously thought.
Collapse
Affiliation(s)
- Mireia Duñach
- a Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina , Universitat Autònoma de Barcelona , Bellaterra , Spain
| | - Beatriz Del Valle-Pérez
- a Departament de Bioquímica i Biologia Molecular, CEB, Facultat de Medicina , Universitat Autònoma de Barcelona , Bellaterra , Spain
| | - Antonio García de Herreros
- b Programa de Recerca en Càncer , Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) , Barcelona , Spain.,c Departament de Ciències Experimentals i de la Salut , Universitat Pompeu Fabra , Barcelona , Spain
| |
Collapse
|
23
|
Marciano DK. A holey pursuit: lumen formation in the developing kidney. Pediatr Nephrol 2017; 32:7-20. [PMID: 26902755 PMCID: PMC5495142 DOI: 10.1007/s00467-016-3326-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/23/2015] [Accepted: 01/12/2016] [Indexed: 02/07/2023]
Abstract
The formation of polarized epithelial tubules is a hallmark of kidney development. One of the fundamental principles in tubulogenesis is that epithelia coordinate the polarity of individual cells with the surrounding cells and matrix. A central feature in this process is the segregation of membranes into spatially and functionally distinct apical and basolateral domains, and the generation of a luminal space at the apical surface. This review examines our current understanding of the cellular and molecular mechanisms that underlie the establishment of apical-basal polarity and lumen formation in developing renal epithelia, including the roles of cell-cell and cell-matrix interactions and polarity complexes. We highlight growing evidence from animal models, and correlate these findings with models of tubulogenesis from other organ systems, and from in vitro studies.
Collapse
Affiliation(s)
- Denise K. Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. H5.102, Dallas, TX 75390-8856
| |
Collapse
|
24
|
Six2creFrs2α knockout mice are a novel model of renal cystogenesis. Sci Rep 2016; 6:36736. [PMID: 27853247 PMCID: PMC5113122 DOI: 10.1038/srep36736] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/14/2016] [Indexed: 01/06/2023] Open
Abstract
Six2cre-mediated deletion of Frs2α (Six2creFrs2αKO), a major fibroblast growth factor receptor (Fgfr) docking protein in mouse nephron progenitors results in perinatal renal hypoplasia; however, postnatal Six2creFrs2αKO kidneys develop cysts. We sought to determine the pathogenesis of Six2creFrs2αKO cyst formation. We performed histological assays, Western blots, and quantitative PCR (qPCR). While embryonic day (E) 18.5 Six2Frs2αKO kidneys were hypoplastic and not cystic, postnatal day (P) 7 mutants had proximal tubular-derived cysts that nearly replaced the renal parenchyma by P21. Mutants had high proximal tubular proliferation rates and interstitial fibrosis, similar to known polycystic kidney disease (PKD) models. Six2creFrs2αKO kidneys also had upregulation of Wnt/βcatenin signaling, macrophage infiltration and chemokine production (e.g. ectopic Ccl2 in non-dilated proximal tubules), and augmented hedgehog signaling, features also seen in other PKD models. We saw increased Gli1 (hedgehog readout) in postnatal Six2creFrs2αKO interstitium and ectopic sonic hedgehog (Shh) in subsets of non-dilated P7 mutant proximal tubules (likely driving the stromal Gli expression). As ectopic tubular Shh and Ccl2 expression is seen after acute kidney injury (AKI), we interrogated another bone fide AKI marker, Kim1 and noted ectopic expression in P7 non-dilated proximal tubules. These observations suggest that aberrantly activated “AKI” pathways may drive pathogenesis in PKD.
Collapse
|
25
|
Cadwell CM, Su W, Kowalczyk AP. Cadherin tales: Regulation of cadherin function by endocytic membrane trafficking. Traffic 2016; 17:1262-1271. [PMID: 27624909 DOI: 10.1111/tra.12448] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022]
Abstract
Cadherins are the primary adhesion molecules in adherens junctions and desmosomes and play essential roles in embryonic development. Although significant progress has been made in understanding cadherin structure and function, we lack a clear vision of how cells confer plasticity upon adhesive junctions to allow for cellular rearrangements during development, wound healing and metastasis. Endocytic membrane trafficking has emerged as a fundamental mechanism by which cells confer a dynamic state to adhesive junctions. Recent studies indicate that the juxtamembrane domain of classical cadherins contains multiple endocytic motifs, or "switches," that can be used by cellular membrane trafficking machinery to regulate adhesion. The cadherin-binding protein p120-catenin (p120) appears to be the master regulator of access to these switches, thereby controlling cadherin endocytosis and turnover. This review focuses on p120 and other cadherin-binding proteins, ubiquitin ligases, and growth factors as key modulators of cadherin membrane trafficking.
Collapse
Affiliation(s)
- Chantel M Cadwell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Wenji Su
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Biochemistry, Cell, and Developmental Biology Graduate Training Program, Emory University, Atlanta, Georgia
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
26
|
Inactivation of p120 catenin in mice disturbs intrahepatic bile duct development and aggravates liver carcinogenesis. Eur J Cell Biol 2016; 95:574-584. [PMID: 27769530 DOI: 10.1016/j.ejcb.2016.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/06/2016] [Accepted: 10/09/2016] [Indexed: 11/21/2022] Open
Abstract
p120 catenin (p120ctn) is required for the stability of classic cadherins at the cell surface and is thought to play a central role in modulating cell-cell adhesion. Cytoplasmic p120ctn promotes cell motility, and probably other activities, by modulating the activities of RhoA, Rac and Cdc42. E-cadherin is expressed in periportal but not in perivenous hepatocytes. In contrast, all hepatocytes of normal mouse liver express N-cadherin. Cholangiocytes express exclusively E-cadherin. Mice with p120ctn ablation in hepatocytes and cholangiocytes (p120LiKO mice) were generated by Cre-loxP technology. Livers were examined by histological, immunohistochemical, ultrastructural and serum analysis to determine the effect of the p120ctn ablation on liver structure and function. Mouse hepatocyte differentiation and homeostasis were not impaired. However, hepatoblasts differentiated abnormally into hybrid hepato-biliary cells, ductal plate structures were irregular in p120LiKO newborns, and further development of intrahepatic bile ducts was severely impaired. In adults, enrichment of ductular structures was accompanied by portal inflammation and fibrosis. p120LiKO mice did not spontaneously develop hepatocellular carcinoma but initiation of hepatocarcinogenesis by diethylnitrosamine was accelerated. In summary: p120ctn has a critical role in biliary differentiation and is a potent suppressor of liver tumor growth.
Collapse
|
27
|
Yang Z, Zimmerman SE, Tsunezumi J, Braitsch C, Trent C, Bryant DM, Cleaver O, González-Manchón C, Marciano DK. Role of CD34 family members in lumen formation in the developing kidney. Dev Biol 2016; 418:66-74. [PMID: 27542690 DOI: 10.1016/j.ydbio.2016.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 11/25/2022]
Abstract
Previous studies have shown CD34 family member Podocalyxin is required for epithelial lumen formation in vitro. We demonstrate that Endoglycan, a CD34 family member with homology to Podocalyxin, is produced prior to lumen formation in developing nephrons. Endoglycan localizes to Rab11-containing vesicles in nephron progenitors, and then relocalizes to the apical surface as progenitors epithelialize. Once an apical/luminal surface is formed, Endoglycan (and the actin-binding protein Ezrin) localize to large, intraluminal structures that may be vesicles/exosomes. We generated mice lacking Endoglycan and found mutants had timely initiation of lumen formation and continuous lumens, similar to controls. Mice with conditional deletion of both Endoglycan and Podocalyxin in developing nephrons also had normal tubular lumens. Despite this, Endoglycan/Podocalyxin is required for apical recruitment of the adaptor protein NHERF1, but not Ezrin, in podocyte precursors, a subset of the epithelia. In summary, while CD34 family members appear dispensable for lumen formation, our data identify Endoglycan as a novel pre-luminal marker and suggest lumen formation occurs via vesicular trafficking of apical cargo that includes Endoglycan.
Collapse
Affiliation(s)
- Zhufeng Yang
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Susan E Zimmerman
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Tsunezumi
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Caitlin Braitsch
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cary Trent
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David M Bryant
- Cancer Research UK (CRUK) Beatson Institute, and Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Consuelo González-Manchón
- Centre of Biological Research, CIB-CSIC, Madrid, Spain; CIBER of Rare Diseases, ISCIII, Madrid, Spain
| | - Denise K Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
28
|
Akchurin O, Du Z, Ramkellawan N, Dalal V, Han SH, Pullman J, Müsch A, Susztak K, Reidy KJ. Partitioning-Defective 1a/b Depletion Impairs Glomerular and Proximal Tubule Development. J Am Soc Nephrol 2016; 27:3725-3737. [PMID: 27185860 DOI: 10.1681/asn.2014111124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The kidney is a highly polarized epithelial organ that develops from undifferentiated mesenchyme, although the mechanisms that regulate the development of renal epithelial polarity are incompletely understood. Partitioning-defective 1 (Par1) proteins have been implicated in cell polarity and epithelial morphogenesis; however, the role of these proteins in the developing kidney has not been established. Therefore, we studied the contribution of Par1a/b to renal epithelial development. We examined the renal phenotype of newborn compound mutant mice carrying only one allele of Par1a or Par1b. Loss of three out of four Par1a/b alleles resulted in severe renal hypoplasia, associated with impaired ureteric bud branching. Compared with kidneys of newborn control littermates, kidneys of newborn mutant mice exhibited dilated proximal tubules and immature glomeruli, and the renal proximal tubular epithelia lacked proper localization of adhesion complexes. Furthermore, Par1a/b mutants expressed low levels of renal Notch ligand Jag1, activated Notch2, and Notch effecter Hes1. Together, these data demonstrate that Par1a/b has a key role in glomerular and proximal tubule development, likely via modulation of Notch signaling.
Collapse
Affiliation(s)
- Oleh Akchurin
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Zhongfang Du
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Nadira Ramkellawan
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Vidhi Dalal
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Seung Hyeok Han
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James Pullman
- Department of Pathology, Montefiore Medical Center, Bronx, New York; and
| | - Anne Müsch
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kimberly J Reidy
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York; .,Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Hendley AM, Wang YJ, Polireddy K, Alsina J, Ahmed I, Lafaro KJ, Zhang H, Roy N, Savidge SG, Cao Y, Hebrok M, Maitra A, Reynolds AB, Goggins M, Younes M, Iacobuzio-Donahue CA, Leach SD, Bailey JM. p120 Catenin Suppresses Basal Epithelial Cell Extrusion in Invasive Pancreatic Neoplasia. Cancer Res 2016; 76:3351-63. [PMID: 27032419 DOI: 10.1158/0008-5472.can-15-2268] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 03/14/2016] [Indexed: 01/01/2023]
Abstract
Aberrant regulation of cellular extrusion can promote invasion and metastasis. Here, we identify molecular requirements for early cellular invasion using a premalignant mouse model of pancreatic cancer with conditional knockout of p120 catenin (Ctnnd1). Mice with biallelic loss of p120 catenin progressively develop high-grade pancreatic intraepithelial neoplasia (PanIN) lesions and neoplasia accompanied by prominent acute and chronic inflammatory processes, which is mediated, in part, through NF-κB signaling. Loss of p120 catenin in the context of oncogenic Kras also promotes remarkable apical and basal epithelial cell extrusion. Abundant single epithelial cells exit PanIN epithelium basally, retain epithelial morphology, survive, and display features of malignancy. Similar extrusion defects are observed following p120 catenin knockdown in vitro, and these effects are completely abrogated by the activation of S1P/S1pr2 signaling. In the context of oncogenic Kras, p120 catenin loss significantly reduces expression of genes mediating S1P/S1pr2 signaling in vivo and in vitro, and this effect is mediated at least, in part, through activation of NF-κB. These results provide insight into mechanisms controlling early events in the metastatic process and suggest that p120 catenin and S1P/S1pr2 signaling enhance cancer progression by regulating epithelial cell invasion. Cancer Res; 76(11); 3351-63. ©2016 AACR.
Collapse
Affiliation(s)
- Audrey M Hendley
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland. The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland. Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Yue J Wang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland. The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kishore Polireddy
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Janivette Alsina
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ishrat Ahmed
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kelly J Lafaro
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland. The David Rubenstein Pancreatic Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Nilotpal Roy
- Diabetes Center, University of California, San Francisco, San Francisco, California
| | - Samuel G Savidge
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland. The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yanna Cao
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Matthias Hebrok
- Diabetes Center, University of California, San Francisco, San Francisco, California
| | - Anirban Maitra
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland. The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Departments of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael Goggins
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland. The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mamoun Younes
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, Texas
| | - Christine A Iacobuzio-Donahue
- The David Rubenstein Pancreatic Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland. The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven D Leach
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland. The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland. The David Rubenstein Pancreatic Cancer Research Center, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Jennifer M Bailey
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland. The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland. Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
30
|
Yu HH, Dohn MR, Markham NO, Coffey RJ, Reynolds AB. p120-catenin controls contractility along the vertical axis of epithelial lateral membranes. J Cell Sci 2015; 129:80-94. [PMID: 26585313 DOI: 10.1242/jcs.177550] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/12/2015] [Indexed: 12/26/2022] Open
Abstract
In vertebrate epithelia, p120-catenin (hereafter referred to as p120; also known as CTNND1) mediates E-cadherin stability and suppression of RhoA. Genetic ablation of p120 in various epithelial tissues typically causes striking alterations in tissue function and morphology. Although these effects could very well involve p120's activity towards Rho, ascertaining the impact of this relationship has been complicated by the fact that p120 is also required for cell-cell adhesion. Here, we have molecularly uncoupled p120's cadherin-stabilizing and RhoA-suppressing activites. Unexpectedly, removing p120's Rho-suppressing activity dramatically disrupted the integrity of the apical surface, irrespective of E-cadherin stability. The physical defect was tracked to excessive actomyosin contractility along the vertical axis of lateral membranes. Thus, we suggest that p120's distinct activities towards E-cadherin and Rho are molecularly and functionally coupled and this, in turn, enables the maintenance of cell shape in the larger context of an epithelial monolayer. Importantly, local suppression of contractility by cadherin-bound p120 appears to go beyond regulating cell shape, as loss of this activity also leads to major defects in epithelial lumenogenesis.
Collapse
Affiliation(s)
- Huapeng H Yu
- Department of Cancer Biology, Vanderbilt University, 37232 Nashville, TN, USA
| | - Michael R Dohn
- Department of Cancer Biology, Vanderbilt University, 37232 Nashville, TN, USA Department of Pharmacology, Vanderbilt University, 37232 Nashville, TN, USA
| | - Nicholas O Markham
- Department of Cancer Biology, Vanderbilt University, 37232 Nashville, TN, USA School of Medicine, Vanderbilt University, 37232 Nashville, TN, USA
| | - Robert J Coffey
- School of Medicine, Vanderbilt University, 37232 Nashville, TN, USA
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University, 37232 Nashville, TN, USA
| |
Collapse
|
31
|
Hong JY, Oh IH, McCrea PD. Phosphorylation and isoform use in p120-catenin during development and tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:102-14. [PMID: 26477567 DOI: 10.1016/j.bbamcr.2015.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/12/2022]
Abstract
P120-catenin is essential to vertebrate development, modulating cadherin and small-GTPase functions, and growing evidence points also to roles in the nucleus. A complexity in addressing p120-catenin's functions is its many isoforms, including optional splicing events, alternative points of translational initiation, and secondary modifications. In this review, we focus upon how choices in the initiation of protein translation, or the earlier splicing of the RNA transcript, relates to primary sequences that harbor established or putative regulatory phosphorylation sites. While certain p120 phosphorylation events arise via known kinases/phosphatases and have defined outcomes, in most cases the functional consequences are still to be established. In this review, we provide examples of p120-isoforms as they relate to phosphorylation events, and thereby to isoform dependent protein-protein associations and downstream functions. We also provide a view of upstream pathways that determine p120's phosphorylation state, and that have an impact upon development and disease. Because other members of the p120 subfamily undergo similar processing and phosphorylation, as well as related catenins of the plakophilin subfamily, what is learned regarding p120 will by extension have wide relevance in vertebrates.
Collapse
Affiliation(s)
- Ji Yeon Hong
- Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Republic of Korea.
| | - Il-Hoan Oh
- The Catholic University of Korea, Catholic High Performance Cell Therapy Center, 505 Banpo-dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center, University of Texas Graduate School of Biomedical Science, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Von Stetina SE, Mango SE. PAR-6, but not E-cadherin and β-integrin, is necessary for epithelial polarization in C. elegans. Dev Biol 2015; 403:5-14. [PMID: 25773364 DOI: 10.1016/j.ydbio.2015.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 01/29/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
Cell polarity is a fundamental characteristic of epithelial cells. Classical cell biological studies have suggested that establishment and orientation of polarized epithelia depend on outside-in cues that derive from interactions with either neighboring cells or the substratum (Akhtar and Streuli, 2013; Chen and Zhang, 2013; Chung and Andrew, 2008; McNeill et al., 1990; Nejsum and Nelson, 2007; Nelson et al., 2013; Ojakian and Schwimmer, 1994; Wang et al., 1990; Yu et al., 2005). This paradigm has been challenged by examples of epithelia generated in the absence of molecules that mediate cell-cell or cell-matrix interactions, notably E-cadherin and integrins (Baas et al., 2004; Choi et al., 2013; Costa et al., 1998; Harris and Peifer, 2004; Raich et al., 1999; Roote and Zusman, 1995; Vestweber et al., 1985; Williams and Waterston, 1994; Wu et al., 2009). Here we explore an alternative hypothesis, that cadherins and integrins function redundantly to substitute for one another during epithelium formation (Martinez-Rico et al., 2010; Ojakian et al., 2001; Rudkouskaya et al., 2014; Weber et al., 2011). We use C. elegans, which possesses a single E-cadherin (Costa et al., 1998; Hardin et al., 2013; Tepass, 1999) and a single β-integrin (Gettner et al., 1995; Lee et al., 2001), and analyze the arcade cells, which generate an epithelium late in embryogenesis (Portereiko and Mango, 2001; Portereiko et al., 2004), after most maternal factors are depleted. Loss of E-cadherin(HMR-1) in combination with β-integrin(PAT-3) had no impact on the onset or formation of the arcade cell epithelium, nor the epidermis or digestive tract. Moreover, ß-integrin(PAT-3) was not enriched at the basal surface of the arcades, and the candidate PAT-3 binding partner β-laminin(LAM-1) was not detected until after arcade cell polarity was established and exhibited no obvious polarity defect when mutated. Instead, the polarity protein par-6 (Chen and Zhang, 2013; Watts et al., 1996) was required to polarize the arcade cells, and par-6 mutants exhibited mislocalized or absent apical and junctional proteins. We conclude that the arcade cell epithelium polarizes by a PAR-6-mediated pathway that is independent of E-cadherin, β-integrin and β-laminin.
Collapse
Affiliation(s)
- Stephen E Von Stetina
- Department of Molecular and Cellular Biology, Harvard University, Boston, MA 02138, USA.
| | - Susan E Mango
- Department of Molecular and Cellular Biology, Harvard University, Boston, MA 02138, USA.
| |
Collapse
|
33
|
Zhang D, Tang N, Liu Y, Wang EH. ARVCF expression is significantly correlated with the malignant phenotype of non-small cell lung cancer. Mol Carcinog 2015; 54 Suppl 1:E185-91. [PMID: 25683624 DOI: 10.1002/mc.22281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/08/2014] [Accepted: 12/12/2014] [Indexed: 01/14/2023]
Abstract
Armadillo repeat gene deleted in velo-cardio-facial syndrome (ARVCF) is a member of the p120 catenin (p120ctn) family; it contains nine central Armadillo repeats and binds to the juxtamembrane domain of E-cadherin. We used immunohistochemistry to measure ARVCF expression in 121 patients with NSCLC and western blotting to examine differences in ARVCF expression between lung cancer and adjacent normal lung tissues. We interfered with ARVCF expression in two lung cancer cell lines and measured its effects on invasion and proliferation. ARVCF expression correlated with the malignant phenotype and poor prognosis. We also observed ARVCF-dependent changes in small GTPase (mainly RhoA) activity in lung cancer cells. We confirmed that ARVCF plays an important role in the malignant phenotype.
Collapse
Affiliation(s)
- Di Zhang
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Na Tang
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - Yang Liu
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| | - En-Hua Wang
- The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China
| |
Collapse
|
34
|
Abstract
The mammalian kidney forms via cell-cell interactions between an epithelial outgrowth of the nephric duct and the surrounding nephrogenic mesenchyme. Initial morphogenetic events include ureteric bud branching to form the collecting duct (CD) tree and mesenchymal-to-epithelial transitions to form the nephrons, requiring reciprocal induction between adjacent mesenchyme and epithelial cells. Within the tips of the branching ureteric epithelium, cells respond to mesenchyme-derived trophic factors by proliferation, migration, and mitosis-associated cell dispersal. Self-inhibition signals from one tip to another play a role in branch patterning. The position, survival, and fate of the nephrogenic mesenchyme are regulated by ECM and secreted signals from adjacent tip and stroma. Signals from the ureteric tip promote mesenchyme self-renewal and trigger nephron formation. Subsequent fusion to the CDs, nephron segmentation and maturation, and formation of a patent glomerular basement membrane also require specialized cell-cell interactions. Differential cadherin, laminin, nectin, and integrin expression, as well as intracellular kinesin and actin-mediated regulation of cell shape and adhesion, underlies these cell-cell interactions. Indeed, the capacity for the kidney to form via self-organization has now been established both via the recapitulation of expected morphogenetic interactions after complete dissociation and reassociation of cellular components during development as well as the in vitro formation of 3D kidney organoids from human pluripotent stem cells. As we understand more about how the many cell-cell interactions required for kidney formation operate, this enables the prospect of bioengineering replacement structures based on these self-organizing properties.
Collapse
|
35
|
Hendley AM, Provost E, Bailey JM, Wang YJ, Cleveland MH, Blake D, Bittman RW, Roeser JC, Maitra A, Reynolds AB, Leach SD. p120 Catenin is required for normal tubulogenesis but not epithelial integrity in developing mouse pancreas. Dev Biol 2014; 399:41-53. [PMID: 25523391 DOI: 10.1016/j.ydbio.2014.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/20/2022]
Abstract
The intracellular protein p120 catenin aids in maintenance of cell-cell adhesion by regulating E-cadherin stability in epithelial cells. In an effort to understand the biology of p120 catenin in pancreas development, we ablated p120 catenin in mouse pancreatic progenitor cells, which resulted in deletion of p120 catenin in all epithelial lineages of the developing mouse pancreas: islet, acinar, centroacinar, and ductal. Loss of p120 catenin resulted in formation of dilated epithelial tubules, expansion of ductal epithelia, loss of acinar cells, and the induction of pancreatic inflammation. Aberrant branching morphogenesis and tubulogenesis were also observed. Throughout development, the phenotype became more severe, ultimately resulting in an abnormal pancreas comprised primarily of duct-like epithelium expressing early progenitor markers. In pancreatic tissue lacking p120 catenin, overall epithelial architecture remained intact; however, actin cytoskeleton organization was disrupted, an observation associated with increased cytoplasmic PKCζ. Although we observed reduced expression of adherens junction proteins E-cadherin, β-catenin, and α-catenin, p120 catenin family members p0071, ARVCF, and δ-catenin remained present at cell membranes in homozygous p120(f/f) pancreases, potentially providing stability for maintenance of epithelial integrity during development. Adult mice homozygous for deletion of p120 catenin displayed dilated main pancreatic ducts, chronic pancreatitis, acinar to ductal metaplasia (ADM), and mucinous metaplasia that resembles PanIN1a. Taken together, our data demonstrate an essential role for p120 catenin in pancreas development.
Collapse
Affiliation(s)
- Audrey M Hendley
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Elayne Provost
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Jennifer M Bailey
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Yue J Wang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Megan H Cleveland
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Danielle Blake
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ross W Bittman
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Jeffrey C Roeser
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Anirban Maitra
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Departments of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Steven D Leach
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
36
|
Zhao H, Zhao Y, Jiang G, Zhang X, Zhang Y, Dong Q, Luan L, Papavassiliou P, Wang E, Wang E. Dishevelled-3 activates p65 to upregulate p120-catenin transcription via a p38-dependent pathway in non-small cell lung cancer. Mol Carcinog 2014; 54 Suppl 1:E112-21. [PMID: 25156800 DOI: 10.1002/mc.22196] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/20/2014] [Accepted: 05/27/2014] [Indexed: 12/26/2022]
Abstract
Dishevelled-3 (Dvl-3) and p120-catenin (p120ctn) have abnormal expression in non-small cell lung cancer (NSCLC), which is associated with poor prognosis. Dvl-3 upregulates p120ctn transcription in NSCLC cells, but the mechanism is unknown. Here we transiently transfected Dvl-3 cDNA to NSCLC cells. Dvl-3 transfection is sufficient for induction of p38 signaling. In turn, Dvl-3 induces p38-mediated activation of the p65 so as to facilitate its nuclear translocation. Treatment with SB203580 (p38 inhibitor) or BAY 11-7082 (IκB-α phosphorylation inhibitor) suppresses Dvl-3 induced activation of p65. The results further show that active p65 interacts with PAX2 promoter to increase the expression of PAX2 and then PAX2 binds to p120ctn promoter so as to upregulate p120ctn gene transcription. Moreover, Dvl-3 transfection enhanced the binding of active p65 to Sp1 so as to decrease the binding of Sp1 to p120ctn promoter. The above-mentioned effects are linked to biological behavior of non-small cell lung cancer cells. These findings confirm that p38 and PAX2 are important for the Dvl-3 induced upregulation of p120ctn. Dvl-3 activates a p38 → p65 → PAX2 → p120ctn pathway to affect biological behavior of NSCLC cells.
Collapse
Affiliation(s)
- Huanyu Zhao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| | - Yue Zhao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| | - Guiyang Jiang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| | - Xiupeng Zhang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| | - Yijun Zhang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| | - Qianze Dong
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| | - Lan Luan
- Department of Pathology, Fengtian Hospital Affiliated to Shenyang Medical College, China
| | | | - Endi Wang
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Enhua Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, China
| |
Collapse
|
37
|
Schackmann RCJ, Tenhagen M, van de Ven RAH, Derksen PWB. p120-catenin in cancer - mechanisms, models and opportunities for intervention. J Cell Sci 2014; 126:3515-25. [PMID: 23950111 DOI: 10.1242/jcs.134411] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The epithelial adherens junction is an E-cadherin-based complex that controls tissue integrity and is stabilized at the plasma membrane by p120-catenin (p120, also known as CTNND1). Mutational and epigenetic inactivation of E-cadherin has been strongly implicated in the development and progression of cancer. In this setting, p120 translocates to the cytosol where it exerts oncogenic properties through aberrant regulation of Rho GTPases, growth factor receptor signaling and derepression of Kaiso (also known as ZBTB33) target genes. In contrast, indirect inactivation of the adherens junction through conditional knockout of p120 in mice was recently linked to tumor formation, indicating that p120 can also function as a tumor suppressor. Supporting these opposing functions are findings in human cancer, which show that either loss or cytoplasmic localization of p120 is a common feature in the progression of several types of carcinoma. Underlying this dual biological phenomenon might be the context-dependent regulation of Rho GTPases in the cytosol and the derepression of Kaiso target genes. Here, we discuss past and present findings that implicate p120 in the regulation of cancer progression and highlight opportunities for clinical intervention.
Collapse
Affiliation(s)
- Ron C J Schackmann
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | |
Collapse
|
38
|
Matsumoto S, Fujii S, Sato A, Ibuka S, Kagawa Y, Ishii M, Kikuchi A. A combination of Wnt and growth factor signaling induces Arl4c expression to form epithelial tubular structures. EMBO J 2014; 33:702-718. [PMID: 24562386 PMCID: PMC4000088 DOI: 10.1002/embj.201386942] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/17/2022] Open
Abstract
Growth factor-dependent epithelial morphological changes and proliferation are essential for the formation of tubular structures, but the underlying molecular mechanisms are poorly understood. Co-stimulation with Wnt3a and epidermal growth factor (Wnt3a/EGF) induced development of tubes consisting of intestinal epithelial cells by inducing expression of Arl4c, an Arf-like small GTP-binding protein, in three-dimensional culture, while stimulation with Wnt3a or EGF alone did not. Arl4c expression resulted in rearrangement of the cytoskeleton through activation of Rac and inactivation of Rho properly, which promoted cell growth by inducing nuclear translocation of Yes-associated protein and transcriptional co-activator with PDZ-binding motif (YAP/TAZ) in leading cells. Arl4c was expressed in ureteric bud tips and pretubular structures in the embryonic kidney. In an organoid culture assay, Wnt and fibroblast growth factor signaling simultaneously induced elongation and budding of kidney ureteric buds through Arl4c expression. YAP/TAZ was observed in the nucleus of extending ureteric bud tips. Thus, Arl4c expression induced by a combination of growth factor signaling mechanisms is involved in tube formation.
Collapse
Affiliation(s)
- Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| | - Shinsuke Fujii
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Interdisciplinary Program for Biomedical Sciences (IPBS), Institute for Academic Initiatives, Osaka University, Graduate School of MedicineOsaka, Japan
| | - Akira Sato
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| | - Souji Ibuka
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Department of Pediatric Surgery, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| | - Yoshinori Kagawa
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Japan Science and Technology Agency (JST), CRESTTokyo, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Japan Science and Technology Agency (JST), CRESTTokyo, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| |
Collapse
|
39
|
Kowalczyk AP, Nanes BA. Adherens junction turnover: regulating adhesion through cadherin endocytosis, degradation, and recycling. Subcell Biochem 2014; 60:197-222. [PMID: 22674073 DOI: 10.1007/978-94-007-4186-7_9] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adherens junctions are important mediators of intercellular adhesion, but they are not static structures. They are regularly formed, broken, and rearranged in a variety of situations, requiring changes in the amount of cadherins, the main adhesion molecule in adherens junctions, present at the cell surface. Thus, endocytosis, degradation, and recycling of cadherins are crucial for dynamic regulation of adherens junctions and control of intercellular adhesion. In this chapter, we review the involvement of cadherin endocytosis in development and disease. We discuss the various endocytic pathways available to cadherins, the adaptors involved, and the sorting of internalized cadherin for recycling or lysosomal degradation. In addition, we review the regulatory pathways controlling cadherin endocytosis and degradation, including regulation of cadherin endocytosis by catenins, cadherin ubiquitination, and growth factor receptor signaling pathways. Lastly, we discuss the proteolytic cleavage of cadherins at the plasma membrane.
Collapse
Affiliation(s)
- Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, 30332, Atlanta, GA, USA,
| | | |
Collapse
|
40
|
Stefanatos RK, Bauer C, Vidal M. p120 catenin is required for the stress response in Drosophila. PLoS One 2013; 8:e83942. [PMID: 24349561 PMCID: PMC3861524 DOI: 10.1371/journal.pone.0083942] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 11/19/2013] [Indexed: 11/18/2022] Open
Abstract
p120ctn is a ubiquitously expressed core component of cadherin junctions and essential for vertebrate development. Surprisingly, Drosophila p120ctn (dp120ctn) is dispensable for adherens junctions and development, which has discouraged Drosophila researchers from further pursuing the biological role of dp120ctn. Here we demonstrate that dp120ctn loss results in increased heat shock sensitivity and reduced animal lifespan, which are completely rescued by ectopic expression of a dp120ctn-GFP transgene. Transcriptomic analysis revealed multiple relish/NF-κB target genes differentially expressed upon loss of dp120ctn. Importantly, this aberrant gene expression was rescued by overexpression of dp120ctn-GFP or heterozygosity for relish. Our results uncover a novel role for dp120ctn in the regulation of animal stress response and immune signalling. This may represent an ancient role of p120ctn and can influence further studies in Drosophila and mammals.
Collapse
Affiliation(s)
- Rhoda K. Stefanatos
- Drosophila Approaches to Cancer Laboratory, The Beatson Institute for Cancer Research, Glasgow, Scotland, United Kingdom
| | - Christin Bauer
- Drosophila Approaches to Cancer Laboratory, The Beatson Institute for Cancer Research, Glasgow, Scotland, United Kingdom
| | - Marcos Vidal
- Drosophila Approaches to Cancer Laboratory, The Beatson Institute for Cancer Research, Glasgow, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
41
|
EMT in developmental morphogenesis. Cancer Lett 2013; 341:9-15. [DOI: 10.1016/j.canlet.2013.02.037] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 02/14/2013] [Accepted: 02/14/2013] [Indexed: 12/24/2022]
|
42
|
Peglion F, Etienne-Manneville S. p120catenin alteration in cancer and its role in tumour invasion. Philos Trans R Soc Lond B Biol Sci 2013; 368:20130015. [PMID: 24062585 DOI: 10.1098/rstb.2013.0015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Since its discovery in 1989 as a substrate of the Src oncogene, p120catenin has been revealed as an important player in cancer initiation and tumour dissemination. p120catenin regulates a wide range of cellular processes such as cell-cell adhesion, cell polarity and cell proliferation and plays a pivotal role in morphogenesis, inflammation and innate immunity. The pleiotropic effects of p120catenin rely on its interactions with numerous partners such as classical cadherins at the plasma membrane, Rho-GTPases and microtubules in the cytosol and transcriptional modulators in the nucleus. Alterations of p120catenin in cancer not only concern its expression level but also its intracellular localization and can lead to both pro-invasive and anti-invasive effects. This review focuses on the p120catenin-mediated pathways involved in cell migration and invasion and discusses the potential consequences of major cancer-related p120catenin alterations with respect to tumour spread.
Collapse
Affiliation(s)
- Florent Peglion
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur - CNRS URA 2582, , 25 rue du Dr Roux, 75724 Paris cedex 15, France
| | | |
Collapse
|
43
|
Abstract
Interconnection of epithelial tubules is a crucial process during organogenesis. Organisms have evolved sets of molecular and cellular strategies to generate an interconnected tubular network during animal development. Spatiotemporal control of common cellular strategies includes dissolution of the basement membrane, apoptosis, rearrangements of cell adhesion junctions, and mesenchymal-like invasive cellular behaviors prior to tubular interconnection. Different model systems exhibit varying degrees of active invasive-like behaviors that precede tubular interconnection, which may reflect changes in cell polarity or differential adhesive cell states. Studies in this newly-emerging field of tubular interconnections will provide a greater understanding of pediatric diseases and cancer metastasis, as well as generate fundamentally new insights into lumen formation pathology, or lumopathies.
Collapse
Affiliation(s)
- Robert M Kao
- Departments of Molecular and Cellular Biology and Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
44
|
Yang Z, Zimmerman S, Brakeman PR, Beaudoin GM, Reichardt LF, Marciano DK. De novo lumen formation and elongation in the developing nephron: a central role for afadin in apical polarity. Development 2013; 140:1774-84. [PMID: 23487309 DOI: 10.1242/dev.087957] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A fundamental process in biology is the de novo formation and morphogenesis of polarized tubules. Although these processes are essential for the formation of multiple metazoan organ systems, little is known about the molecular mechanisms that regulate them. In this study, we have characterized several steps in tubule formation and morphogenesis using the mouse kidney as a model system. We report that kidney mesenchymal cells contain discrete Par3-expressing membrane microdomains that become restricted to an apical domain, coinciding with lumen formation. Once lumen formation has been initiated, elongation occurs by simultaneous extension and additional de novo lumen generation. We demonstrate that lumen formation and elongation require afadin, a nectin adaptor protein implicated in adherens junction formation. Mice that lack afadin in nephron precursors show evidence of Par3-expressing membrane microdomains, but fail to develop normal apical-basal polarity and generate a continuous lumen. Absence of afadin led to delayed and diminished integration of nectin complexes and failure to recruit R-cadherin. Furthermore, we demonstrate that afadin is required for Par complex formation. Together, these results suggest that afadin acts upstream of the Par complex to regulate the integration and/or coalescence of membrane microdomains, thereby establishing apical-basal polarity and lumen formation/elongation during kidney tubulogenesis.
Collapse
Affiliation(s)
- Zhufeng Yang
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Members of the p120-catenin family associate with cadherins and regulate their stability at the plasma membrane. How p120-catenin limits cadherin endocytosis has long remained a mystery. In this issue, Nanes et al. (2012. J. Cell Biol. doi:10.1083/jcb.201205029) identify a conserved acidic motif within cadherins that acts as a physical platform for p120-catenin binding. However, in the absence of p120-catenin, the motif acts as an endocytic signal. These results provide new insight into p120-catenin's role as guardian of intercellular junction dynamics.
Collapse
Affiliation(s)
- Mirna Perez-Moreno
- BBVA Foundation-Cancer Cell Biology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid 28019, Spain.
| | | |
Collapse
|
46
|
Nanes BA, Chiasson-MacKenzie C, Lowery AM, Ishiyama N, Faundez V, Ikura M, Vincent PA, Kowalczyk AP. p120-catenin binding masks an endocytic signal conserved in classical cadherins. ACTA ACUST UNITED AC 2013; 199:365-80. [PMID: 23071156 PMCID: PMC3471230 DOI: 10.1083/jcb.201205029] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
p120 regulates adhesive junction dynamics through binding to a dual-function motif in classical cadherins that alternately serves as a p120-binding interface and an endocytic signal. p120-catenin (p120) binds to the cytoplasmic tails of classical cadherins and inhibits cadherin endocytosis. Although p120 regulation of cadherin internalization is thought to be important for adhesive junction dynamics, the mechanism by which p120 modulates cadherin endocytosis is unknown. In this paper, we identify a dual-function motif in classical cadherins consisting of three highly conserved acidic residues that alternately serve as a p120-binding interface and an endocytic signal. Mutation of this motif resulted in a cadherin variant that was both p120 uncoupled and resistant to endocytosis. In endothelial cells, in which dynamic changes in adhesion are important components of angiogenesis and inflammation, a vascular endothelial cadherin (VE-cadherin) mutant defective in endocytosis assembled normally into cell–cell junctions but potently suppressed cell migration in response to vascular endothelial growth factor. These results reveal the mechanistic basis by which p120 stabilizes cadherins and demonstrate that VE-cadherin endocytosis is crucial for endothelial cell migration in response to an angiogenic growth factor.
Collapse
Affiliation(s)
- Benjamin A Nanes
- Graduate Program in Biochemistry, Cell, and Developmental Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Yasuda T, Fukuda M. Slp2-a controls renal epithelial cell size through regulation of Rap–ezrin signaling independently of Rab27. J Cell Sci 2013; 127:557-70. [DOI: 10.1242/jcs.134056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Slp2-a is a Rab27 effector protein that regulates transport of Rab27-bearing vesicles/organelles via its N-terminal Rab27-binding domain and a phospholipid-binding C2A domain. Here we demonstrate a Rab27-independent function of Slp2-a in the control of renal cell size via a previously uncharacterized C2B domain. We found that by recruiting Rap1GAPs to the plasma membrane of MDCK II cells via the C2B domain Slp2-a inactivates Rap signaling and modulates the size of the cells. Functional ablation of Slp2-a resulted in an increase in the size of MDCK II cells. Drosophila Slp bitesize was found to compensate for the function of Slp2-a in MDCK II cells, thereby indicating that the mechanism of the cell size control by Slps has been evolutionarily conserved. Interestingly, blockade of the activity of ezrin, a downstream target of Rap, with the glucosylceramide synthase inhibitor miglustat effectively inhibited cell spreading of Slp2-a-knockdown cells. We also discovered aberrant expression of Slp2-a and increased activity of ezrin in pcy mice, a model of polycystic kidney disease that is characterized by renal cell spreading. Our findings indicate that Slp2-a controls renal cell size through regulation of Rap–ezrin signaling independently of Rab27.
Collapse
|
48
|
Kurley SJ, Bierie B, Carnahan RH, Lobdell NA, Davis MA, Hofmann I, Moses HL, Muller WJ, Reynolds AB. p120-catenin is essential for terminal end bud function and mammary morphogenesis. Development 2012; 139:1754-64. [PMID: 22461563 DOI: 10.1242/dev.072769] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although p120-catenin (p120) is crucial for E-cadherin function, ablation experiments in epithelial tissues from different organ systems reveal markedly different effects. Here, we examine for the first time the consequences of p120 knockout during mouse mammary gland development. An MMTV-Cre driver was used to target knockout to the epithelium at the onset of puberty. p120 ablation was detected in approximately one-quarter of the nascent epithelium at the forth week post-partum. However, p120 null cells were essentially nonadherent, excluded from the process of terminal end bud (TEB) morphogenesis and lost altogether by week six. This elimination process caused a delay in TEB outgrowth, after which the gland developed normally from cells that had retained p120. Mechanistic studies in vitro indicate that TEB dysfunction is likely to stem from striking E-cadherin loss, failure of cell-cell adhesion and near total exclusion from the collective migration process. Our findings reveal an essential role for p120 in mammary morphogenesis.
Collapse
Affiliation(s)
- Sarah J Kurley
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Minuth WW, Denk L. Interstitial interfaces show marked differences in regenerating tubules, matured tubules, and the renal stem/progenitor cell niche. J Biomed Mater Res A 2012; 100:1115-25. [PMID: 22337593 DOI: 10.1002/jbm.a.34039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 11/29/2011] [Indexed: 11/10/2022]
Abstract
Stem/progenitor cells are promising candidates for the regeneration of parenchyma in acute and chronic renal failure. After an implantation stem/progenitor cells must migrate through the interstitial space to concentrate at the site of damage. However, information is lacking to what extent the interstitial interface is influencing the development of stem/progenitor cells into nephron structures. In consequence, tubule regeneration within an artificial polyester interstitium was analyzed by electron microscopy in comparison with the interstitial interface of matured tubules and the interstitium within the renal stem/progenitor cell niche. The experiments demonstrate that fixation of specimens with glutaraldehyde (GA) is leading in all cases to inconspicuously looking interstitial interfaces. In contrast, fixation of regenerating tubules in GA containing ruthenium red and tannic acid shows a dense network of fibers lining along the basal lamina. In contrast, matured tubules reveal after ruthenium red label an extremely thickened basal lamina, while only a punctate pattern is obtained after tannic acid treatment. Finally, within the renal stem/progenitor cell niche ruthenium red and tannic acid label reveals large amounts of extracellular matrix spanning through the interstitium. Thus, fixation of tissue in GA containing ruthenium red and tannic acid exhibits an unexpectedly regional heterogeneity of the renal interstitial interface. This fact has to be considered for an optimal therapeutic repair of parenchyma, since contacts between stem/progenitor cells with the interstitial interface influence further development.
Collapse
Affiliation(s)
- Will W Minuth
- Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany.
| | | |
Collapse
|
50
|
New Insights into the Regulation of E-cadherin Distribution by Endocytosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 295:63-108. [DOI: 10.1016/b978-0-12-394306-4.00008-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|