1
|
Poh QH, Rai A, Cross J, Greening DW. HB-EGF-loaded nanovesicles enhance trophectodermal spheroid attachment and invasion. Proteomics 2024; 24:e2200145. [PMID: 38214697 DOI: 10.1002/pmic.202200145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024]
Abstract
The ability of trophectodermal cells (outer layer of the embryo) to attach to the endometrial cells and subsequently invade the underlying matrix are critical stages of embryo implantation during successful pregnancy establishment. Extracellular vesicles (EVs) have been implicated in embryo-maternal crosstalk, capable of reprogramming endometrial cells towards a pro-implantation signature and phenotype. However, challenges associated with EV yield and direct loading of biomolecules limit their therapeutic potential. We have previously established generation of cell-derived nanovesicles (NVs) from human trophectodermal cells (hTSCs) and their capacity to reprogram endometrial cells to enhance adhesion and blastocyst outgrowth. Here, we employed a rapid NV loading strategy to encapsulate potent implantation molecules such as HB-EGF (NVHBEGF). We show these loaded NVs elicit EGFR-mediated effects in recipient endometrial cells, activating kinase phosphorylation sites that modulate their activity (AKT S124/129, MAPK1 T185/Y187), and downstream signalling pathways and processes (AKT signal transduction, GTPase activity). Importantly, they enhanced target cell attachment and invasion. The phosphoproteomics and proteomics approach highlight NVHBEGF-mediated short-term signalling patterns and long-term reprogramming capabilities on endometrial cells which functionally enhance trophectodermal-endometrial interactions. This proof-of-concept study demonstrates feasibility in enhancing the functional potency of NVs in the context of embryo implantation.
Collapse
Affiliation(s)
- Qi Hui Poh
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Alin Rai
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jonathon Cross
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Fried S, Gilboa D, Har-Zahav A, Lavrut PM, Du Y, Karjoo S, Russo P, Shamir R, Wells RG, Waisbourd-Zinman O. Extrahepatic cholangiocyte obstruction is mediated by decreased glutathione, Wnt and Notch signaling pathways in a toxic model of biliary atresia. Sci Rep 2020; 10:7599. [PMID: 32371929 PMCID: PMC7200694 DOI: 10.1038/s41598-020-64503-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Biliary atresia is a neonatal liver disease with extrahepatic bile duct obstruction and progressive liver fibrosis. The etiology and pathogenesis of the disease are unknown. We previously identified a plant toxin, biliatresone, responsible for biliary atresia in naturally-occurring animal models, that causes cholangiocyte destruction in in-vitro models. Decreases in reduced glutathione (GSH) mimic the effects of biliatresone, and agents that replenish cellular GSH ameliorate the effects of the toxin. The goals of this study were to define signaling pathways downstream of biliatresone that lead to cholangiocyte destruction and to determine their relationship to GSH. Using cholangiocyte culture and 3D cholangiocyte spheroid cultures, we found that biliatresone and decreases in GSH upregulated RhoU/Wrch1, a Wnt signaling family member, which then mediated an increase in Hey2 in the NOTCH signaling pathway, causing downregulation of the transcription factor Sox17. When these genes were up- or down-regulated, the biliatresone effect on spheroids was phenocopied, resulting in lumen obstruction. Biopsies of patients with biliary atresia demonstrated increased RhoU/Wrch1 and Hey2 expression in cholangiocytes. We present a novel pathway of cholangiocyte injury in a model of biliary atresia, which is relevant to human BA and may suggest potential future therapeutics.
Collapse
Affiliation(s)
- Sophia Fried
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Dafna Gilboa
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Adi Har-Zahav
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Yu Du
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Sara Karjoo
- Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Pierre Russo
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rebecca G Wells
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Orith Waisbourd-Zinman
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel. .,Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States.
| |
Collapse
|
3
|
Porazinski S, Parkin A, Pajic M. Rho-ROCK Signaling in Normal Physiology and as a Key Player in Shaping the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:99-127. [PMID: 32030687 DOI: 10.1007/978-3-030-35582-1_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Rho-ROCK signaling network has a range of specialized functions of key biological importance, including control of essential developmental processes such as morphogenesis and physiological processes including homeostasis, immunity, and wound healing. Deregulation of Rho-ROCK signaling actively contributes to multiple pathological conditions, and plays a major role in cancer development and progression. This dynamic network is critical in modulating the intricate communication between tumor cells, surrounding diverse stromal cells and the matrix, shaping the ever-changing microenvironment of aggressive tumors. In this chapter, we overview the complex regulation of the Rho-ROCK signaling axis, its role in health and disease, and analyze progress made with key approaches targeting the Rho-ROCK pathway for therapeutic benefit. Finally, we conclude by outlining likely future trends and key questions in the field of Rho-ROCK research, in particular surrounding Rho-ROCK signaling within the tumor microenvironment.
Collapse
Affiliation(s)
- Sean Porazinski
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Ashleigh Parkin
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Marina Pajic
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Hodge RG, Ridley AJ. Regulation and functions of RhoU and RhoV. Small GTPases 2020; 11:8-15. [PMID: 29189096 PMCID: PMC6959303 DOI: 10.1080/21541248.2017.1362495] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023] Open
Abstract
Rho GTPases play central roles in a wide variety of cellular processes, including cytoskeletal dynamics, cell adhesion and cell polarity. RhoU and RhoV are Rho GTPases that have some atypical properties compared with classical Rho family members, such as the presence of N- and C-terminal extension regions, unusual GDP/GTP cycling and post-translational modification by palmitoylation but not prenylation. Their activity and localization is regulated by the N-terminal and C-terminal regions, and so far no GEFs or GAPs have been identified for them. Similar to Rac and Cdc42, they interact with PAK serine/threonine kinases, and in the case of PAK4, this interaction leads to RhoU protein stabilization. In cells, RhoU and RhoV alter cell shape and cell adhesion, which probably underlies some of the phenotypes reported for these proteins in vivo, for example in heart development and epithelial morphogenesis. However, the molecular basis for these functions of RhoU and RhoV remains to be characterized.
Collapse
Affiliation(s)
- Richard G. Hodge
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Anne J. Ridley
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| |
Collapse
|
5
|
Zhang Q, Liu Y, Wang H, Ma L, Xia H, Niu J, Sun T, Zhang L. The preventive effects of taurine on neural tube defects through the Wnt/PCP-Jnk-dependent pathway. Amino Acids 2017; 49:1633-1640. [PMID: 28718066 DOI: 10.1007/s00726-017-2462-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/03/2017] [Indexed: 11/30/2022]
Abstract
The aim of this study was to clarify the protective role of taurine in neuronal apoptosis and the role of the Wnt/PCP-Jnk pathway in mediating the preventive effects of taurine on neural tube defects (NTDs). HT-22 cells (a hippocampal neuron cell line) were divided into a control group, a glutamate-induced apoptosis group, and glutamate (4.0 mmol/L) plus low-dose taurine (L; 0.5 mmol/L) and high-dose taurine (H; 2.0 mmol/L) groups. The MTT assay was used to monitor cell proliferation and cell survival. Immunofluorescence and Western blot analyses were used to determine caspase 9 expression. Retinoic acid (RA) induced embryonic NTDs in Kunming mice, thus establishing an NTD model. Pregnant mice were divided into a control group, an RA (30 mg/kg body weight) group, and an RA (30 mg/kg body weight) plus taurine (free drinking of 2 g/L solution) group. Immunohistochemistry and Western blot analyses were used to detect the expression of Dvl, RhoA and phosphorylated (p)-Jnk/Jnk in the embryonic neural tubes. In HT-22 cells, the apoptosis rate was significantly higher and caspase 9 activation was also significantly increased in the glutamate-induced apoptosis group compared to the L and H taurine groups. In the NTD model, the expression levels of Dvl, RhoA, and p-Jnk were significantly higher in the RA group than in the control group, whereas they were significantly reduced in the RA + taurine group. This study suggests that taurine has positive effects on neuronal protection and NTD prevention. Moreover, the Wnt/PCP-Jnk-dependent pathway plays an important role in taurine-mediated prevention of NTDs.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China.
| | - Yang Liu
- Department of Neurosurgery, Qingdao Chengyang People's Hospital, Qingdao, 266106, China
| | - Hui Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Monogenic Disease Research Center for Neurological Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Li Ma
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Hechun Xia
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Jianguo Niu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Tao Sun
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Ningxia Key Laboratory of Cerebrocranial Diseases, No. 804 Shenglinajie, XingQing District, Yinchuan, 750004, NingXia, China
| | - Li Zhang
- Department of Cardiac Function Examination, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
6
|
Yanagihara K, Liu Y, Kanie K, Takayama K, Kokunugi M, Hirata M, Fukuda T, Suga M, Nikawa H, Mizuguchi H, Kato R, Furue MK. Prediction of Differentiation Tendency Toward Hepatocytes from Gene Expression in Undifferentiated Human Pluripotent Stem Cells. Stem Cells Dev 2016; 25:1884-1897. [PMID: 27733097 PMCID: PMC5165660 DOI: 10.1089/scd.2016.0099] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Functional hepatocytes derived from human pluripotent stem cells (hPSCs) have potential as tools for predicting drug-induced hepatotoxicity in the early phases of drug development. However, the propensity of hPSC lines to differentiate into specific lineages is reported to differ. The ability to predict low propensity of hPSCs to differentiate into hepatocytes would facilitate the selection of useful hPSC clones and substantially accelerate development of hPSC-derived hepatocytes for pharmaceutical research. In this study, we compared the expression of genes associated with hepatic differentiation in five hPSC lines including human ES cell line, H9, which is known to differentiate into hepatocytes, and an hPSC line reported with a poor propensity for hepatic differentiation. Genes distinguishing between undifferentiated hPSCs, hPSC-derived hepatoblast-like differentiated cells, and primary human hepatocytes were drawn by two-way cluster analysis. The order of expression levels of genes in undifferentiated hPSCs was compared with that in hPSC-derived hepatoblast-like cells. Three genes were selected as predictors of low propensity for hepatic differentiation. Expression of these genes was investigated in 23 hPSC clones. Review of representative cells by induction of hepatic differentiation suggested that low prediction scores were linked with low hepatic differentiation. Thus, our model using gene expression ranking and bioinformatic analysis could reasonably predict poor differentiation propensity of hPSC lines.
Collapse
Affiliation(s)
- Kana Yanagihara
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| | - Yujung Liu
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| | - Kei Kanie
- 2 Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University , Nagoya, Japan
| | - Kazuo Takayama
- 3 Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan .,4 The Keihanshin Consortium for Fostering the Next Generation of Global Leaders in Research (K-CONNEX), Kyoto University , Kyoto, Japan .,5 Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| | - Minako Kokunugi
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan .,6 Department of Oral Biology & Engineering Integrated Health Sciences, Institute of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Mitsuhi Hirata
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| | - Takayuki Fukuda
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| | - Mika Suga
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| | - Hiroki Nikawa
- 6 Department of Oral Biology & Engineering Integrated Health Sciences, Institute of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Hiroyuki Mizuguchi
- 3 Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University , Osaka, Japan .,5 Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan .,7 Global Center for Medical Engineering and Informatics, Osaka University , Osaka, Japan
| | - Ryuji Kato
- 2 Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University , Nagoya, Japan
| | - Miho K Furue
- 1 Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation , Health and Nutrition, Osaka, Japan
| |
Collapse
|
7
|
Pieters T, Goossens S, Haenebalcke L, Andries V, Stryjewska A, De Rycke R, Lemeire K, Hochepied T, Huylebroeck D, Berx G, Stemmler MP, Wirth D, Haigh JJ, van Hengel J, van Roy F. p120 Catenin-Mediated Stabilization of E-Cadherin Is Essential for Primitive Endoderm Specification. PLoS Genet 2016; 12:e1006243. [PMID: 27556156 PMCID: PMC4996431 DOI: 10.1371/journal.pgen.1006243] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
E-cadherin-mediated cell-cell adhesion is critical for naive pluripotency of cultured mouse embryonic stem cells (mESCs). E-cadherin-depleted mESC fail to downregulate their pluripotency program and are unable to initiate lineage commitment. To further explore the roles of cell adhesion molecules during mESC differentiation, we focused on p120 catenin (p120ctn). Although one key function of p120ctn is to stabilize and regulate cadherin-mediated cell-cell adhesion, it has many additional functions, including regulation of transcription and Rho GTPase activity. Here, we investigated the role of mouse p120ctn in early embryogenesis, mESC pluripotency and early fate determination. In contrast to the E-cadherin-null phenotype, p120ctn-null mESCs remained pluripotent, but their in vitro differentiation was incomplete. In particular, they failed to form cystic embryoid bodies and showed defects in primitive endoderm formation. To pinpoint the underlying mechanism, we undertook a structure-function approach. Rescue of p120ctn-null mESCs with different p120ctn wild-type and mutant expression constructs revealed that the long N-terminal domain of p120ctn and its regulatory domain for RhoA were dispensable, whereas its armadillo domain and interaction with E-cadherin were crucial for primitive endoderm formation. We conclude that p120ctn is not only an adaptor and regulator of E-cadherin, but is also indispensable for proper lineage commitment.
Collapse
Affiliation(s)
- Tim Pieters
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Steven Goossens
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lieven Haenebalcke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Vanessa Andries
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Agata Stryjewska
- Department of Development and Regeneration, Laboratory of Molecular Biology (Celgen), University of Leuven, Leuven, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Kelly Lemeire
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Tino Hochepied
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
| | - Danny Huylebroeck
- Department of Development and Regeneration, Laboratory of Molecular Biology (Celgen), University of Leuven, Leuven, Belgium
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Geert Berx
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Marc P. Stemmler
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Freiburg, Germany
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Dagmar Wirth
- Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Jody J. Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Alfred Centre, Melbourne, Victoria, Australia
| | - Jolanda van Hengel
- Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- * E-mail: (JvH); (FvR)
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Inflammation Research Center, VIB, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- * E-mail: (JvH); (FvR)
| |
Collapse
|
8
|
Loebel DAF, Plageman TF, Tang TL, Jones VJ, Muccioli M, Tam PPL. Thyroid bud morphogenesis requires CDC42- and SHROOM3-dependent apical constriction. Biol Open 2016; 5:130-9. [PMID: 26772200 PMCID: PMC4823982 DOI: 10.1242/bio.014415] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Early development of the gut endoderm and its subsequent remodeling for the formation of organ buds are accompanied by changes to epithelial cell shape and polarity. Members of the Rho-related family of small GTPases and their interacting proteins play multiple roles in regulating epithelial morphogenesis. In this study we examined the role of Cdc42 in foregut development and organ bud formation. Ablation of Cdc42 in post-gastrulation mouse embryos resulted in a loss of apical-basal cell polarity and columnar epithelial morphology in the ventral pharyngeal endoderm, in conjunction with a loss of apical localization of the known CDC42 effector protein PARD6B. Cell viability but not proliferation in the foregut endoderm was impaired. Outgrowth of the liver, lung and thyroid buds was severely curtailed in Cdc42-deficient embryos. In particular, the thyroid bud epithelium did not display the apical constriction that normally occurs concurrently with the outgrowth of the bud into the underlying mesenchyme. SHROOM3, a protein that interacts with Rho GTPases and promotes apical constriction, was strongly expressed in the thyroid bud and its sub-cellular localization was disrupted in Cdc42-deficient embryos. In Shroom3 gene trap mutant embryos, the thyroid bud epithelium showed no apical constriction, while the bud continued to grow and protruded into the foregut lumen. Our findings indicate that Cdc42 is required for epithelial polarity and organization in the endoderm and for apical constriction in the thyroid bud. It is possible that the function of CDC42 is partly mediated by SHROOM3. Summary: Conditional Cdc42 knockout revealed requirements for Cdc42 in endoderm polarity, and in thyroid apical constriction and morphogenesis. Shroom3 mutant embryos also displayed thyroid bud abnormalities, suggesting a possible functional interaction.
Collapse
Affiliation(s)
- David A F Loebel
- Embryology Unit, Children's Medical Research Institute, Locked Bag 32, Wentworthville, New South Wales 2145, Australia Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Timothy F Plageman
- Ohio State University College of Optometry, Columbus, OH 43210-1280, USA
| | - Theresa L Tang
- Embryology Unit, Children's Medical Research Institute, Locked Bag 32, Wentworthville, New South Wales 2145, Australia
| | - Vanessa J Jones
- Embryology Unit, Children's Medical Research Institute, Locked Bag 32, Wentworthville, New South Wales 2145, Australia
| | - Maria Muccioli
- Ohio State University College of Optometry, Columbus, OH 43210-1280, USA
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, Locked Bag 32, Wentworthville, New South Wales 2145, Australia Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
9
|
Mesenchymal-epithelial interactions during digestive tract development and epithelial stem cell regeneration. Cell Mol Life Sci 2015; 72:3883-96. [PMID: 26126787 DOI: 10.1007/s00018-015-1975-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 12/16/2022]
Abstract
The gastrointestinal tract develops from a simple and uniform tube into a complex organ with specific differentiation patterns along the anterior-posterior and dorso-ventral axes of asymmetry. It is derived from all three germ layers and their cross-talk is important for the regulated development of fetal and adult gastrointestinal structures and organs. Signals from the adjacent mesoderm are essential for the morphogenesis of the overlying epithelium. These mesenchymal-epithelial interactions govern the development and regionalization of the different gastrointestinal epithelia and involve most of the key morphogens and signaling pathways, such as the Hedgehog, BMPs, Notch, WNT, HOX, SOX and FOXF cascades. Moreover, the mechanisms underlying mesenchyme differentiation into smooth muscle cells influence the regionalization of the gastrointestinal epithelium through interactions with the enteric nervous system. In the neonatal and adult gastrointestinal tract, mesenchymal-epithelial interactions are essential for the maintenance of the epithelial regionalization and digestive epithelial homeostasis. Disruption of these interactions is also associated with bowel dysfunction potentially leading to epithelial tumor development. In this review, we will discuss various aspects of the mesenchymal-epithelial interactions observed during digestive epithelium development and differentiation and also during epithelial stem cell regeneration.
Collapse
|
10
|
RNA-sequencing analysis reveals new alterations in cardiomyocyte cytoskeletal genes in patients with heart failure. J Transl Med 2014; 94:645-53. [PMID: 24709777 DOI: 10.1038/labinvest.2014.54] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/12/2014] [Accepted: 02/26/2014] [Indexed: 12/15/2022] Open
Abstract
Changes in cardiomyocyte cytoskeletal components, a crucial scaffold of cellular structure, have been found in heart failure (HF); however, the altered cytoskeletal network remains to be elucidated. This study investigated a new map of cytoskeleton-linked alterations that further explain the cardiomyocyte morphology and contraction disruption in HF. RNA-Sequencing (RNA-Seq) analysis was performed in 29 human LV tissue samples from ischemic cardiomyopathy (ICM; n=13) and dilated cardiomyopathy (DCM, n=10) patients undergoing cardiac transplantation and six healthy donors (control, CNT) and up to 16 ICM, 13 DCM and 7 CNT tissue samples for qRT-PCR. Gene Ontology analysis of RNA-Seq data demonstrated that cytoskeletal processes are altered in HF. We identified 60 differentially expressed cytoskeleton-related genes in ICM and 58 genes in DCM comparing with CNT, hierarchical clustering determined that shared cytoskeletal genes have a similar behavior in both pathologies. We further investigated MYLK4, RHOU, and ANKRD1 cytoskeletal components. qRT-PCR analysis revealed that MYLK4 was downregulated (-2.2-fold; P<0.05) and ANKRD1 was upregulated (2.3-fold; P<0.01) in ICM patients vs CNT. RHOU mRNA levels showed a statistical trend to decrease (-2.9-fold). In DCM vs CNT, MYLK4 (-4.0-fold; P<0.05) and RHOU (-3.9-fold; P<0.05) were downregulated and ANKRD1 (2.5-fold; P<0.05) was upregulated. Accordingly, MYLK4 and ANKRD1 protein levels were decreased and increased, respectively, in both diseases. Furthermore, ANKRD1 and RHOU mRNA levels were related with LV function (P<0.05). In summary, we have found a new map of changes in the ICM and DCM cardiomyocyte cytoskeleton. ANKRD1 and RHOU mRNA levels were related with LV function which emphasizes their relevance in HF. These new cytoskeletal changes may be responsible for altered contraction and cell architecture disruption in HF patients. Moreover, these results improve our knowledge on the role of cytoskeleton in functional and structural alterations in HF.
Collapse
|