1
|
Phillips AT, Boumil EF, Venkatesan A, Tilstra-Smith C, Castro N, Knox BE, Henty-Ridilla JL, Bernstein AM. The formin DAAM1 regulates the deubiquitinase activity of USP10 and integrin homeostasis. Eur J Cell Biol 2023; 102:151347. [PMID: 37562219 PMCID: PMC10839120 DOI: 10.1016/j.ejcb.2023.151347] [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: 07/12/2022] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
The differentiation of fibroblasts into pathological myofibroblasts during wound healing is characterized by increased cell surface expression of αv-integrins. Our previous studies found that the deubiquitinase (DUB) USP10 removes ubiquitin from αv-integrins, leading to cell surface integrin accumulation, subsequent TGFβ1 activation, and pathological myofibroblast differentiation. In this study, a yeast two-hybrid screen revealed a novel binding partner for USP10, the formin, DAAM1. We found that DAAM1 binds to and inhibits USP10's DUB activity through the FH2 domain of DAAM1 independent of its actin functions. The USP10/DAAM1 interaction was also supported by proximity ligation assay (PLA) in primary human corneal fibroblasts. Treatment with TGFβ1 significantly increased USP10 and DAAM1 protein expression, PLA signal, and co-localization to actin stress fibers. DAAM1 siRNA knockdown significantly reduced co-precipitation of USP10 and DAAM1 on purified actin stress fibers, and β1- and β5-integrin ubiquitination. This resulted in increased αv-, β1-, and β5-integrin total protein levels, αv-integrin recycling, and extracellular fibronectin (FN) deposition. Together, our data demonstrate that DAAM1 inhibits USP10's DUB activity on integrins subsequently regulating cell surface αv-integrin localization and FN accumulation.
Collapse
Affiliation(s)
- Andrew T Phillips
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Edward F Boumil
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Arunkumar Venkatesan
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Christine Tilstra-Smith
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Nileyma Castro
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA
| | - Barry E Knox
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Jessica L Henty-Ridilla
- SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Audrey M Bernstein
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA.
| |
Collapse
|
2
|
Gao L, Qiu F, Cao H, Li H, Dai G, Ma T, Gong Y, Luo W, Zhu D, Qiu Z, Zhu P, Chu S, Yang H, Liu Z. Therapeutic delivery of microRNA-125a-5p oligonucleotides improves recovery from myocardial ischemia/reperfusion injury in mice and swine. Theranostics 2023; 13:685-703. [PMID: 36632217 PMCID: PMC9830430 DOI: 10.7150/thno.73568] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 12/12/2022] [Indexed: 01/04/2023] Open
Abstract
Rationale: Clinical application of mesenchymal stem cells (MSCs) and MSC-derived exosomes (MSC-Exos) to alleviate myocardial ischemia/reperfusion (I/R) injury is compromised by the low cell engraftment rate and uncontrolled exosomal content. As one of their active ingredients, single-component microRNA therapy may have more inherent advantages. We sought to find an ideal microRNA candidate and determine whether it could reproduce the cardioprotective effects of MSCs and MSC-Exos. Methods: Cardiac function and myocardial remodeling in MSC, MSC-Exo, or microRNA oligonucleotide-treated mouse hearts were investigated after I/R injury. The effects of microRNA oligonucleotides on cardiac cells (macrophages, cardiomyocytes, fibroblasts, and endothelial cells) and their downstream mechanisms were confirmed. Large animals were also employed to investigate the safety of microRNA therapy. Results: The results showed that microRNA-125a-5p (miR-125a-5p) is enriched in MSC-Exos, and intramyocardial delivery of their modified oligonucleotides (agomir) in mouse I/R myocardium, as well as MSCs or MSC-Exos, exerted obvious cardioprotection by increasing cardiac function and limiting adverse remodeling. In addition, miR-125a-5p agomir treatment increased M2 macrophage polarization, promoted angiogenesis, and attenuated fibroblast proliferation and activation, which subsequently contributed to the improvements in cardiomyocyte apoptosis and inflammation. Mechanistically, Klf13, Tgfbr1, and Daam1 are considered the targets of miR-125a-5p for regulating the function of macrophages, fibroblasts, and endothelial cells, respectively. Similar results were observed following miR-125a-5p agomir treatment in a porcine model, with no increase in the risk of arrhythmia or hepatic, renal, or cardiac toxicity. Conclusions: This targeted microRNA delivery presents an effective and safe strategy as a stem cell and exosomal therapy in I/R cardiac repair.
Collapse
Affiliation(s)
- Ling Gao
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| | - Fan Qiu
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Hao Cao
- Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Hao Li
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Gonghua Dai
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Teng Ma
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Yanshan Gong
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Wei Luo
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Dongling Zhu
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Zhixuan Qiu
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Shuguang Chu
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| | - Huangtian Yang
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China.,Research Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai 200031, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China.,✉ Corresponding authors: Ling Gao, PhD, Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 1800 Yuntai Rd., Shanghai, 200123, China. E-mail: ; Zhongmin Liu, MD, PhD, Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd., Shanghai, 200120, China. E-mail: ; Huangtian Yang, PhD, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, 320 Yueyang Rd., Shanghai, 200031, China. E-mail: ; Shuguang Chu, PhD, Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China. E-mail:
| |
Collapse
|
3
|
Sun BB, Loomis SJ, Pizzagalli F, Shatokhina N, Painter JN, Foley CN, Jensen ME, McLaren DG, Chintapalli SS, Zhu AH, Dixon D, Islam T, Ba Gari I, Runz H, Medland SE, Thompson PM, Jahanshad N, Whelan CD. Genetic map of regional sulcal morphology in the human brain from UK biobank data. Nat Commun 2022; 13:6071. [PMID: 36241887 PMCID: PMC9568560 DOI: 10.1038/s41467-022-33829-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 10/05/2022] [Indexed: 12/24/2022] Open
Abstract
Genetic associations with macroscopic brain structure can provide insights into brain function and disease. However, specific associations with measures of local brain folding are largely under-explored. Here, we conducted large-scale genome- and exome-wide associations of regional cortical sulcal measures derived from magnetic resonance imaging scans of 40,169 individuals in UK Biobank. We discovered 388 regional brain folding associations across 77 genetic loci, with genes in associated loci enriched for expression in the cerebral cortex, neuronal development processes, and differential regulation during early brain development. We integrated brain eQTLs to refine genes for various loci, implicated several genes involved in neurodevelopmental disorders, and highlighted global genetic correlations with neuropsychiatric phenotypes. We provide an interactive 3D visualisation of our summary associations, emphasising added resolution of regional analyses. Our results offer new insights into the genetic architecture of brain folding and provide a resource for future studies of sulcal morphology in health and disease.
Collapse
Affiliation(s)
- Benjamin B Sun
- Translational Biology, Research & Development, Biogen Inc., Cambridge, MA, US.
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Stephanie J Loomis
- Translational Biology, Research & Development, Biogen Inc., Cambridge, MA, US
| | - Fabrizio Pizzagalli
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US
| | - Natalia Shatokhina
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US
| | - Jodie N Painter
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christopher N Foley
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Optima Partners, Edinburgh, UK
| | - Megan E Jensen
- Clinical Sciences, Research & Development, Biogen Inc., Cambridge, MA, US
| | - Donald G McLaren
- Clinical Sciences, Research & Development, Biogen Inc., Cambridge, MA, US
| | | | - Alyssa H Zhu
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US
| | - Daniel Dixon
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US
| | - Tasfiya Islam
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US
| | - Iyad Ba Gari
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US
| | - Heiko Runz
- Translational Biology, Research & Development, Biogen Inc., Cambridge, MA, US
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US.
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, US.
| | | |
Collapse
|
4
|
Wang W, Shang W, Zou J, Liu K, Liu M, Qiu X, Zhang H, Wang K, Wang N. ZNF667 facilitates angiogenesis after myocardial ischemia through transcriptional regulation of VASH1 and Wnt signaling pathway. Int J Mol Med 2022; 50:129. [PMID: 36043524 PMCID: PMC9448299 DOI: 10.3892/ijmm.2022.5185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
Zinc finger protein 667 (ZNF667, also referred as Mipu1), a widely expressed KRAB/C2H2-type zinc finger transcription factor, can protect against hypoxic-ischemic myocardial injury. Pro-angiogenesis is regarded as a promising strategy for the treatment of acute myocardial infarction (AMI). However, whether ZNF667 is involved in the angiogenesis following AMI remains to be elucidated. The present study reported that the expression of ZNF667 in CD31-positive endothelial cells (ECs) was upregulated in the heart of AMI mice. Hypoxic challenge (1% oxygen) promoted the mRNA and protein expression of ZNF667 in the human umbilical vein endothelial cells (HUVECs) in a time-dependent manner. Moreover, ZNF667 promoted hypoxia-induced invasion and tube formation of HUVECs. Mechanically, ZNF667 could directly bind to the promoter of anti-angiogenic gene VASH1 and inhibit its expression. Consequently, VASH1 overexpression abolished hypoxic challenge or ZNF667 overexpression-induced invasion and tube formation of HUVECs. Further bioinformatic analyses suggested that overexpression of ZNF667 or knockdown of VASH1-induced differentially expressed genes in HUVECs were greatly enriched in the Wnt signaling pathway (DAAM1, LEF1, RAC2, FRAT1, NFATc2 and WNT5A). Together, these data suggested that ZNF667 facilitates myocardial ischemia-driven angiogenesis through transcriptional repression of VASH1 and regulation of Wnt signaling pathway.
Collapse
Affiliation(s)
- Wenmei Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weite Shang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jiang Zou
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaoqin Qiu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
5
|
Ahangar P, Cowin AJ. Reforming the Barrier: The Role of Formins in Wound Repair. Cells 2022; 11:cells11182779. [PMID: 36139355 PMCID: PMC9496773 DOI: 10.3390/cells11182779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/04/2022] Open
Abstract
The restoration of an intact epidermal barrier after wound injury is the culmination of a highly complex and exquisitely regulated physiological process involving multiple cells and tissues, overlapping dynamic events and protein synthesis and regulation. Central to this process is the cytoskeleton, a system of intracellular proteins that are instrumental in regulating important processes involved in wound repair including chemotaxis, cytokinesis, proliferation, migration, and phagocytosis. One highly conserved family of cytoskeletal proteins that are emerging as major regulators of actin and microtubule nucleation, polymerization, and stabilization are the formins. The formin family includes 15 different proteins categorized into seven subfamilies based on three formin homology domains (FH1, FH2, and FH3). The formins themselves are regulated in different ways including autoinhibition, activation, and localization by a range of proteins, including Rho GTPases. Herein, we describe the roles and effects of the formin family of cytoskeletal proteins on the fundamental process of wound healing and highlight recent advances relating to their important functions, mechanisms, and regulation at the molecular and cellular levels.
Collapse
|
6
|
Wang C, Qu K, Wang J, Qin R, Li B, Qiu J, Wang G. Biomechanical regulation of planar cell polarity in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166495. [PMID: 35850177 DOI: 10.1016/j.bbadis.2022.166495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2023]
Abstract
Cell polarity refers to the uneven distribution of certain cytoplasmic components in a cell with a spatial order. The planar cell polarity (PCP), the cell aligns perpendicular to the polar plane, in endothelial cells (ECs) has become a research hot spot. The planar polarity of ECs has a positive significance on the regulation of cardiovascular dysfunction, pathological angiogenesis, and ischemic stroke. The endothelial polarity is stimulated and regulated by biomechanical force. Mechanical stimuli promote endothelial polarization and make ECs produce PCP to maintain the normal physiological and biochemical functions. Here, we overview recent advances in understanding the interplay and mechanism between PCP and ECs function involved in mechanical forces, with a focus on PCP signaling pathways and organelles in regulating the polarity of ECs. And then showed the related diseases caused by ECs polarity dysfunction. This study provides new ideas and therapeutic targets for the treatment of endothelial PCP-related diseases.
Collapse
Affiliation(s)
- Caihong Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Rui Qin
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Bingyi Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| |
Collapse
|
7
|
Wang J, Feng D, Gao B. An Overview of Potential Therapeutic Agents Targeting WNT/PCP Signaling. Handb Exp Pharmacol 2021; 269:175-213. [PMID: 34463852 DOI: 10.1007/164_2021_533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Since the discovery of the proto-oncogene Wnt1 (Int1) in 1982, WNT signaling has been identified as one of the most important pathways that regulates a wide range of fundamental developmental and physiological processes in multicellular organisms. The canonical WNT signaling pathway depends on the stabilization and translocation of β-catenin and plays important roles in development and homeostasis. The WNT/planar cell polarity (WNT/PCP) signaling, also known as one of the β-catenin-independent WNT pathways, conveys directional information to coordinate polarized cell behaviors. Similar to WNT/β-catenin signaling, disruption or aberrant activation of WNT/PCP signaling also underlies a variety of developmental defects and cancers. However, the pharmacological targeting of WNT/PCP signaling for therapeutic purposes remains largely unexplored. In this review, we briefly discuss WNT/PCP signaling in development and disease and summarize the known drugs/inhibitors targeting this pathway.
Collapse
Affiliation(s)
- Jin Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Di Feng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
8
|
Wang G, Lin F, Wan Q, Wu J, Luo M. Mechanisms of action of metformin and its regulatory effect on microRNAs related to angiogenesis. Pharmacol Res 2020; 164:105390. [PMID: 33352227 DOI: 10.1016/j.phrs.2020.105390] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023]
Abstract
Angiogenesis is rapidly initiated in response to pathological conditions and is a key target for pharmaceutical intervention in various malignancies. Anti-angiogenic therapy has emerged as a potential and effective therapeutic strategy for treating cancer and cardiovascular-related diseases. Metformin, a first-line oral antidiabetic agent for type 2 diabetes mellitus (T2DM), not only reduces blood glucose levels and improves insulin sensitivity and exerts cardioprotective effects but also shows benefits against cancers, cardiovascular diseases, and other diverse diseases and regulates angiogenesis. MicroRNAs (miRNAs) are endogenous noncoding RNA molecules with a length of approximately 19-25 bases that are widely involved in controlling various human biological processes. A large number of miRNAs are involved in the regulation of cardiovascular cell function and angiogenesis, of which miR-21 not only regulates vascular cell proliferation, migration and apoptosis but also plays an important role in angiogenesis. The relationship between metformin and abnormal miRNA expression has gradually been revealed in the context of numerous diseases and has received increasing attention. This paper reviews the drug-target interactions and drug repositioning events of metformin that influences vascular cells and has benefits on angiogenesis-mediated effects. Furthermore, we use miR-21 as an example to explain the specific molecular mechanism underlying metformin-mediated regulation of the miRNA signaling pathway controlling angiogenesis and vascular protective effects. These findings may provide a new therapeutic target and theoretical basis for the clinical prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Fang Lin
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Qin Wan
- Department of Endocrinology, Nephropathy Clinical Medical Research Center of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
9
|
Vaeyens MM, Jorge-Peñas A, Barrasa-Fano J, Shapeti A, Roeffaers M, Van Oosterwyck H. Actomyosin-dependent invasion of endothelial sprouts in collagen. Cytoskeleton (Hoboken) 2020; 77:261-276. [PMID: 32588525 DOI: 10.1002/cm.21624] [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: 04/16/2020] [Revised: 06/11/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
During sprouting angiogenesis-the growth of blood vessels from the existing vasculature-endothelial cells (ECs) adopt an elongated invasive form and exert forces at cell-cell and cell-matrix interaction sites. These cell shape changes and cellular tractions require extensive reorganizations of the actomyosin network. However, the respective roles of actin and myosin for endothelial sprouting are not fully elucidated. In this study, we further investigate these roles by treating 2D-migrating and 3D-sprouting ECs with chemical compounds targeting either myosin or actin. These treatments affected the endothelial cytoskeleton drastically and reduced the invasive response in a compound-specific manner; pointing toward a tight control of the actin and myosin activity during sprouting. Clusters in the data further illustrate that endothelial sprout morphology is sensitive to the in vitro model mechanical microenvironment and directs future research toward mechanical substrate guidance as a strategy for promoting engineered tissue vascularization. In summary, our results add to a growing corpus of research highlighting a key role of the cytoskeleton for sprouting angiogenesis.
Collapse
Affiliation(s)
- Marie-Mo Vaeyens
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alvaro Jorge-Peñas
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jorge Barrasa-Fano
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Apeksha Shapeti
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Roeffaers
- Department of Microbial and Molecular Systems (M2S), Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Nakaya MA, Gudmundsson KO, Komiya Y, Keller JR, Habas R, Yamaguchi TP, Ajima R. Placental defects lead to embryonic lethality in mice lacking the Formin and PCP proteins Daam1 and Daam2. PLoS One 2020; 15:e0232025. [PMID: 32353019 PMCID: PMC7192421 DOI: 10.1371/journal.pone.0232025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 04/06/2020] [Indexed: 01/30/2023] Open
Abstract
The actin cytoskeleton plays a central role in establishing cell polarity and shape during embryonic morphogenesis. Daam1, a member of the Formin family of actin cytoskeleton regulators, is a Dvl2-binding protein that functions in the Wnt/Planar Cell Polarity (PCP) pathway. To examine the role of the Daam proteins in mammalian development, we generated Daam-deficient mice by gene targeting and found that Daam1, but not Daam2, is necessary for fetal survival. Embryonic development of Daam1 mutants was delayed most likely due to functional defects in the labyrinthine layer of the placenta. Examination of Daam2 and Daam1/2 double mutants revealed that Daam1 and Daam2 are functionally redundant during placental development. Of note, neural tube closure defects (NTD), which are observed in several mammalian PCP mutants, are not observed in Wnt5a or Daam1 single mutants, but arise in Daam1;Wnt5a double mutants. These findings demonstrate a unique function for Daam genes in placental development and are consistent with a role for Daam1 in the Wnt/PCP pathway in mammals.
Collapse
Affiliation(s)
- Masa-aki Nakaya
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland, United State of America
| | - Kristibjorn Orri Gudmundsson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland, United State of America
| | - Yuko Komiya
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United State of America
| | - Jonathan R. Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland, United State of America
| | - Raymond Habas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United State of America
| | - Terry P. Yamaguchi
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland, United State of America
| | - Rieko Ajima
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland, United State of America
| |
Collapse
|
11
|
Lee M, Hwang YS, Yoon J, Sun J, Harned A, Nagashima K, Daar IO. Developmentally regulated GTP-binding protein 1 modulates ciliogenesis via an interaction with Dishevelled. J Cell Biol 2019; 218:2659-2676. [PMID: 31270137 PMCID: PMC6683737 DOI: 10.1083/jcb.201811147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/25/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
Our study reveals Drg1 as a new binding partner of Dishevelled. The Drg1–Dishevelled association regulates Daam1 and RhoA interactions and activity, leading to polymerization and stability of the actin cytoskeleton, a process that is essential for proper multiciliation. Cilia are critical for proper embryonic development and maintaining homeostasis. Although extensively studied, there are still significant gaps regarding the proteins involved in regulating ciliogenesis. Using the Xenopus laevis embryo, we show that Dishevelled (Dvl), a key Wnt signaling scaffold that is critical to proper ciliogenesis, interacts with Drg1 (developmentally regulated GTP-binding protein 1). The loss of Drg1 or disruption of the interaction with Dvl reduces the length and number of cilia and displays defects in basal body migration and docking to the apical surface of multiciliated cells (MCCs). Moreover, Drg1 morphants display abnormal rotational polarity of basal bodies and a decrease in apical actin and RhoA activity that can be attributed to disruption of the protein complex between Dvl and Daam1, as well as between Daam1 and RhoA. These results support the concept that the Drg1–Dvl interaction regulates apical actin polymerization and stability in MCCs. Thus, Drg1 is a newly identified partner of Dvl in regulating ciliogenesis.
Collapse
Affiliation(s)
| | | | - Jaeho Yoon
- National Cancer Institute, Frederick, MD
| | - Jian Sun
- National Cancer Institute, Frederick, MD
| | - Adam Harned
- Electron Microscope Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kunio Nagashima
- Electron Microscope Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Ira O Daar
- National Cancer Institute, Frederick, MD
| |
Collapse
|
12
|
Abstract
Formin homology proteins (formins) are a highly conserved family of cytoskeletal remodeling proteins that are involved in a diverse array of cellular functions. Formins are best known for their ability to regulate actin dynamics, but the same functional domains also govern stability and organization of microtubules. It is thought that this dual activity allows them to coordinate the activity of these two major cytoskeletal networks and thereby influence cellular architecture. Golgi ribbon assembly is dependent upon cooperative interactions between actin filaments and cytoplasmic microtubules originating both at the Golgi itself and from the centrosome. Similarly, centrosome assembly, centriole duplication, and centrosome positioning are also reliant on a dialogue between both cytoskeletal networks. As presented in this chapter, a growing body of evidence suggests that multiple formin proteins play essential roles in these central cellular processes.
Collapse
Affiliation(s)
- John Copeland
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
13
|
Wang Z, Liu CH, Huang S, Chen J. Wnt Signaling in vascular eye diseases. Prog Retin Eye Res 2018; 70:110-133. [PMID: 30513356 DOI: 10.1016/j.preteyeres.2018.11.008] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/21/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022]
Abstract
The Wnt signaling pathway plays a pivotal role in vascular morphogenesis in various organs including the eye. Wnt ligands and receptors are key regulators of ocular angiogenesis both during the eye development and in vascular eye diseases. Wnt signaling participates in regulating multiple vascular beds in the eye including regression of the hyaloid vessels, and development of structured layers of vasculature in the retina. Loss-of-function mutations in Wnt signaling components cause rare genetic eye diseases in humans such as Norrie disease, and familial exudative vitreoretinopathy (FEVR) with defective ocular vasculature. On the other hand, experimental studies in more prevalent vascular eye diseases, such as wet age-related macular degeneration (AMD), diabetic retinopathy (DR), retinopathy of prematurity (ROP), and corneal neovascularization, suggest that aberrantly increased Wnt signaling is one of the causations for pathological ocular neovascularization, indicating the potential of modulating Wnt signaling to ameliorate pathological angiogenesis in eye diseases. This review recapitulates the key roles of the Wnt signaling pathway during ocular vascular development and in vascular eye diseases, and pharmaceutical approaches targeting the Wnt signaling as potential treatment options.
Collapse
Affiliation(s)
- Zhongxiao Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Shuo Huang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States.
| |
Collapse
|
14
|
Fan J, Ray P, Lu Y, Kaur G, Schwarz JJ, Wan LQ. Cell chirality regulates intercellular junctions and endothelial permeability. SCIENCE ADVANCES 2018; 4:eaat2111. [PMID: 30397640 PMCID: PMC6200360 DOI: 10.1126/sciadv.aat2111] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/13/2018] [Indexed: 06/08/2023]
Abstract
Cell chirality is a newly discovered intrinsic property of the cell, reflecting the bias of the cell to polarize in the left-right axis. Despite increasing evidence on its substantial role in the asymmetric development of embryos, little is known about implications of cell chirality in physiology and disease. We demonstrate that cell chirality accounts for the nonmonotonic, dose-response relationship between endothelial permeability and protein kinase C (PKC) activation. The permeability of the endothelial cell layer is tightly controlled in our body, and dysregulation often leads to tissue inflammation and diseases. Our results show that low-level PKC activation is sufficient to reverse cell chirality through phosphatidylinositol 3-kinase/AKT signaling and alters junctional protein organization between cells with opposite chirality, leading to an unexpected substantial change in endothelial permeability. Our findings suggest that cell chirality regulates intercellular junctions in important ways, providing new opportunities for drug delivery across tightly connected semipermeable cellular sheets.
Collapse
Affiliation(s)
- Jie Fan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Poulomi Ray
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Yaowei Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Gurleen Kaur
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - John J. Schwarz
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
15
|
Zeng YF, Xiao YS, Liu Y, Luo XJ, Wen LD, Liu Q, Chen M. Formin-like 3 regulates RhoC/FAK pathway and actin assembly to promote cell invasion in colorectal carcinoma. World J Gastroenterol 2018; 24:3884-3897. [PMID: 30228782 PMCID: PMC6141330 DOI: 10.3748/wjg.v24.i34.3884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/16/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To clarify the underlying mechanism of formin-like 3 (FMNL3) in the promotion of colorectal carcinoma (CRC) cell invasion.
METHODS The in vitro biological function analyses of FMNL3 were performed by gain- and loss-of function approaches. Changes in the F-actin cytoskeleton were detected by the technologies of phalloidin-TRITC labeling and confocal microscopy. The signaling pathway mediated by FMNL3 was explored by western blot, gelatin zymograph assay, co-immunoprecipitation (co-IP), immunofluorescence co-localization, and glutathione S-transferase (GST) pull-down assay.
RESULTS The in vitro experimental results showed that FMNL3 significantly promoted the proliferation, invasion, and migration of CRC cells (P < 0.05 and P < 0.01). Moreover, FMNL3 regulated the remodeling of actin-based protrusions such as filopodia and lamellipodia in a RhoC-dependent manner. The western blot and gelatin zymograph assay results indicated that FMNL3 was involved in the RhoC/ focal adhesion kinase (FAK) pathway and acted as an effector of RhoC to activate the downstream signaling of p-FAK as well as p-MAPK and p-AKT. This resulted in the increased expression of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9) and vascular endothelial growth factor (VEGF), and the subsequent promotion of CRC cell invasion. The results of TAE226, U0126 or Ly294002 treatment confirmed an essential role of FMNL3 in activation of the RhoC/FAK pathway and the subsequent promotion of CRC invasion. Co-IP, co-localization and GST pull-down assays showed the direct interaction of FMNL3 with RhoC in vivo and in vitro.
CONCLUSION FMNL3 regulates the RhoC/FAK signaling pathway and RhoC-dependent remodeling of actin-based protrusions to promote CRC invasion.
Collapse
Affiliation(s)
- Yuan-Feng Zeng
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Yi-Sheng Xiao
- Teaching and Researching Section of Morphology, College of Basic Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yong Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Xiao-Jiang Luo
- Department of General Surgery, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Li-Dan Wen
- Clinical Medical Sciences Institute, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Qian Liu
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| | - Min Chen
- Department of Pathology, Jiangxi Provincial People’s Hospital, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
16
|
The Role of scaRNAs in Adjusting Alternative mRNA Splicing in Heart Development. J Cardiovasc Dev Dis 2018; 5:jcdd5020026. [PMID: 29738469 PMCID: PMC6023535 DOI: 10.3390/jcdd5020026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 12/19/2022] Open
Abstract
Congenital heart disease (CHD) is a leading cause of death in children <1 year of age. Despite intense effort in the last 10 years, most CHDs (~70%) still have an unknown etiology. Conotruncal based defects, such as Tetralogy of Fallot (TOF), a common complex of devastating heart defects, typically requires surgical intervention in the first year of life. We reported that the noncoding transcriptome in myocardial tissue from children with TOF is characterized by significant variation in levels of expression of noncoding RNAs, and more specifically, a significant reduction in 12 small cajal body-associated RNAs (scaRNAs) in the right ventricle. scaRNAs are essential for the biochemical modification and maturation of small nuclear RNAs (spliceosomal RNAs), which in turn are critical components of the spliceosome. This is particularly important because we also documented that splicing of mRNAs that are critical for heart development was dysregulated in the heart tissue of infants with TOF. Furthermore, we went on to show, using the zebrafish model, that altering the expression of these same scaRNAs led to faulty mRNA processing and heart defects in the developing embryo. This review will examine how scaRNAs may influence spliceosome fidelity in exon retention during heart development and thus contribute to regulation of heart development.
Collapse
|
17
|
Venditti M, Fasano C, Santillo A, Aniello F, Minucci S. First evidence of DAAM1 localization in mouse seminal vesicles and its possible involvement during regulated exocytosis. C R Biol 2018; 341:228-234. [PMID: 29571963 DOI: 10.1016/j.crvi.2018.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/01/2018] [Accepted: 03/01/2018] [Indexed: 12/26/2022]
Abstract
Dishevelled-associated activator of morphogenesis 1 (DAAM1) is a protein belonging to the formin family, which regulates, together with the small GTPase RhoA, the nucleation and the assembly of actin fibres through Wnt-Dishevelled PCP pathway. Its role has been investigated in essential biological processes, such as cell polarity, movement and adhesion during morphogenesis and organogenesis. In this work, we studied the expression of DAAM1 mRNA and protein by PCR and Western blot analyses and its co-localization with actin in adult mouse seminal vesicles by immunofluorescence. We show that both proteins are cytoplasmic: actin is evident at cell-cell junctions and at cell cortex; DAAM1 had a more diffused localization, but is also prominent at the apical plasmatic membrane of epithelial cells. These findings support our hypothesis of a role of DAAM1 in cytoskeletal rearrangement that occurs during the exocytosis of secretory vesicles, and in particular concerning actin filaments. We were also able to detect DAAM1 and actin association in the smooth muscle cells that surround the epithelium too. In this case, we could only speculate the possible involvement of this formin in muscular cells in the maintenance and the regulation of the contractile structures. The present results strongly suggest that DAAM1 could have a pivotal role in vesicle exocytosis and in the physiology of mouse seminal vesicles.
Collapse
Affiliation(s)
- Massimo Venditti
- Dipartimento di Medicina Sperimentale, Sez, Fisiologia Umana e Funzioni Biologiche Integrate, Università degli studi della Campania "Luigi Vanvitelli", Via Costantinopoli, 16, 80138 Napoli, Italy
| | - Chiara Fasano
- Dipartimento di Medicina Sperimentale, Sez, Fisiologia Umana e Funzioni Biologiche Integrate, Università degli studi della Campania "Luigi Vanvitelli", Via Costantinopoli, 16, 80138 Napoli, Italy
| | - Alessandra Santillo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Francesco Aniello
- Dipartimento di Biologia, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Sergio Minucci
- Dipartimento di Medicina Sperimentale, Sez, Fisiologia Umana e Funzioni Biologiche Integrate, Università degli studi della Campania "Luigi Vanvitelli", Via Costantinopoli, 16, 80138 Napoli, Italy.
| |
Collapse
|
18
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
19
|
Salman MM, Kitchen P, Woodroofe MN, Bill RM, Conner AC, Heath PR, Conner MT. Transcriptome Analysis of Gene Expression Provides New Insights into the Effect of Mild Therapeutic Hypothermia on Primary Human Cortical Astrocytes Cultured under Hypoxia. Front Cell Neurosci 2017; 11:386. [PMID: 29311824 PMCID: PMC5735114 DOI: 10.3389/fncel.2017.00386] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/20/2017] [Indexed: 01/02/2023] Open
Abstract
Hypothermia is increasingly used as a therapeutic measure to treat brain injury. However, the cellular mechanisms underpinning its actions are complex and are not yet fully elucidated. Astrocytes are the most abundant cell type in the brain and are likely to play a critical role. In this study, transcriptional changes and the protein expression profile of human primary cortical astrocytes cultured under hypoxic conditions for 6 h were investigated. Cells were treated either with or without a mild hypothermic intervention 2 h post-insult to mimic the treatment of patients following traumatic brain injury (TBI) and/or stroke. Using human gene expression microarrays, 411 differentially expressed genes were identified following hypothermic treatment of astrocytes following a 2 h hypoxic insult. KEGG pathway analysis indicated that these genes were mainly enriched in the Wnt and p53 signaling pathways, which were inhibited following hypothermic intervention. The expression levels of 168 genes involved in Wnt signaling were validated by quantitative real-time-PCR (qPCR). Among these genes, 10 were up-regulated and 32 were down-regulated with the remainder unchanged. Two of the differentially expressed genes (DEGs), p38 and JNK, were selected for validation at the protein level using cell based ELISA. Hypothermic intervention significantly down-regulated total protein levels for the gene products of p38 and JNK. Moreover, hypothermia significantly up-regulated the phosphorylated (activated) forms of JNK protein, while downregulating phosphorylation of p38 protein. Within the p53 signaling pathway, 35 human apoptosis-related proteins closely associated with Wnt signaling were investigated using a Proteome Profiling Array. Hypothermic intervention significantly down-regulated 18 proteins, while upregulating one protein, survivin. Hypothermia is a complex intervention; this study provides the first detailed longitudinal investigation at the transcript and protein expression levels of the molecular effects of therapeutic hypothermic intervention on hypoxic human primary cortical astrocytes. The identified genes and proteins are targets for detailed functional studies, which may help to develop new treatments for brain injury based on an in-depth mechanistic understanding of the astrocytic response to hypoxia and/or hypothermia.
Collapse
Affiliation(s)
- Mootaz M Salman
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Philip Kitchen
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - M Nicola Woodroofe
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Roslyn M Bill
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Alex C Conner
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Matthew T Conner
- Research Institute of Health Sciences, Wolverhampton School of Sciences, University of Wolverhampton, Wolverhampton, United Kingdom
| |
Collapse
|
20
|
Fu SW, Chen HY, Lin XL, Yang L, Ge ZZ. Collagen triple helix repeat containing 1 promotes tumor angiogenesis in gastrointestinal stromal tumors. Oncol Lett 2017; 14:7499-7505. [PMID: 29344195 DOI: 10.3892/ol.2017.7111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 11/25/2016] [Indexed: 12/13/2022] Open
Abstract
Collagen triple helix repeat containing 1 (Cthrc1) is a secreted protein that has been observed to lead to poorer prognosis by inducing the invasion and metastasis in different tumors; however, it has not been demonstrated that Cthrc1 is involved in tumor angiogenesis. Immunohistochemical staining of Cthrc1 and CD31 in gastrointestinal stromal tumor tissue demonstrated that Cthrc1 is associated with microvascular density. Overexpression of Cthrc1 protein may alter the properties of human umbilical vein endothelial cells (HUVECs), including migration, invasion, tubule formation and aortic ring sprouting. Small interfering RNA-mediated knockdown of Cthrc1 was performed to verify the opposite effects. Migration and tubule formation induced by Cthrc1 overexpression in HUVECs was attenuated by inhibition of phosphorylation in extracellular-signal-regulated protein kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling pathways. The pro-angiogenic effect of Cthcr1 is associated with increased phosphorylation of ERK and JNK in HUVECs. Silencing the expression of Cthrc1 protein may be a promising strategy to inhibit tumor angiogenesis.
Collapse
Affiliation(s)
- Seng Wang Fu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, P.R. China
| | - Hai Ying Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, P.R. China
| | - Xiao Lu Lin
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, P.R. China
| | - Li Yang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, P.R. China
| | - Zhi Zheng Ge
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, P.R. China
| |
Collapse
|
21
|
Lu Y, Zhang Y, Pan MH, Kim NH, Sun SC, Cui XS. Daam1 regulates fascin for actin assembly in mouse oocyte meiosis. Cell Cycle 2017; 16:1350-1356. [PMID: 28682694 DOI: 10.1080/15384101.2017.1325045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
As a formin protein, Daam1 (Dishevelled-associated activator of morphogenesis 1) is reported to regulate series of cell processes like endocytosis, cell morphology and migration via its effects on actin assembly in mitosis. However, whether Daam1 plays roles in female meiosis remains uncertain. In this study, we investigated the expression and functions of Daam1 during mouse oocyte meiosis. Our results indicated that Daam1 localized at the cortex of oocytes, which was similar with actin filaments. After Daam1 morpholino (MO) microinjection, the expression of Daam1 significantly decreased, which resulted in the failure of oocyte polar body extrusion. These results might be due to the defects of actin assembly, since the decreased fluorescence intensity of actin filaments in oocyte cortex and cytoplasm were observed. However, Daam1 knockdown seemed not to affect the meiotic spindle movement. In addition, we found that fascin might be the down effector of Daam1, since the protein expression of fascin decreased after Daam1 knockdown. Thus, our data suggested that Daam1 affected actin assembly during oocyte meiotic division via the regulation of fascin expression.
Collapse
Affiliation(s)
- Yujie Lu
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Yu Zhang
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Meng-Hao Pan
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Nam-Hyung Kim
- b Department of Animal Sciences , Chungbuk National University , Cheongju , Korea
| | - Shao-Chen Sun
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Xiang-Shun Cui
- b Department of Animal Sciences , Chungbuk National University , Cheongju , Korea
| |
Collapse
|
22
|
Manichaikul A, Wang XQ, Sun L, Dupuis J, Borczuk AC, Nguyen JN, Raghu G, Hoffman EA, Onengut-Gumuscu S, Farber EA, Kaufman JD, Rabinowitz D, Stukovsky KDH, Kawut SM, Hunninghake GM, Washko GR, O'Connor GT, Rich SS, Barr RG, Lederer DJ. Genome-wide association study of subclinical interstitial lung disease in MESA. Respir Res 2017; 18:97. [PMID: 28521775 PMCID: PMC5437638 DOI: 10.1186/s12931-017-0581-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 05/10/2017] [Indexed: 12/14/2022] Open
Abstract
Background We conducted a genome-wide association study (GWAS) of subclinical interstitial lung disease (ILD), defined as high attenuation areas (HAA) on CT, in the population-based Multi-Ethnic Study of Atherosclerosis Study. Methods We measured the percentage of high attenuation areas (HAA) in the lung fields on cardiac CT scan defined as voxels with CT attenuation values between -600 and -250 HU. Genetic analyses were performed in MESA combined across race/ethnic groups: non-Hispanic White (n = 2,434), African American (n = 2,470), Hispanic (n = 2,065) and Chinese (n = 702), as well as stratified by race/ethnicity. Results Among 7,671 participants, regions at genome-wide significance were identified for basilar peel-core ratio of HAA in FLJ35282 downstream of ANRIL (rs7852363, P = 2.1x10−9) and within introns of SNAI3-AS1 (rs140142658, P = 9.6x10−9) and D21S2088E (rs3079677, P = 2.3x10−8). Within race/ethnic groups, 18 additional loci were identified at genome-wide significance, including genes related to development (FOXP4), cell adhesion (ALCAM) and glycosylation (GNPDA2, GYPC, GFPT1 and FUT10). Among these loci, SNP rs6844387 near GNPDA2 demonstrated nominal evidence of replication in analysis of n = 1,959 participants from the Framingham Heart Study (P = 0.029). FOXP4 region SNP rs2894439 demonstrated evidence of validation in analysis of n = 228 White ILD cases from the Columbia ILD Study compared to race/ethnicity-matched controls from MESA (one-sided P = 0.007). In lung tissue from 15 adults with idiopathic pulmonary fibrosis compared to 15 adults without lung disease. ANRIL (P = 0.001), ALCAM (P = 0.03) and FOXP4 (P = 0.046) were differentially expressed. Conclusions Our results suggest novel roles for protein glycosylation and cell cycle disinhibition by long non-coding RNA in the pathogenesis of ILD. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0581-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA. .,Department of Public Health Sciences, Biostatistics Section, University of Virginia, Charlottesville, VA, USA. .,Center for Public Health Genomics, University of Virginia School of Medicine, West Complex Room 6115, Charlottesville, VA, 22903, USA.
| | - Xin-Qun Wang
- Department of Public Health Sciences, Biostatistics Section, University of Virginia, Charlottesville, VA, USA
| | - Li Sun
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.,The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Alain C Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, NY, USA
| | - Jennifer N Nguyen
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Ganesh Raghu
- University of Washington Center for Interstitial Lung Diseases, Seattle, WA, USA
| | - Eric A Hoffman
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Emily A Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Joel D Kaufman
- Departmenst of Environmental & Occupational Health Sciences, Medicine, and Epidemiology, University of Washington, Seattle, WA, USA
| | - Dan Rabinowitz
- Department of Statistics, Columbia University, New York, NY, USA
| | | | - Steven M Kawut
- Department of Medicine and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gary M Hunninghake
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George T O'Connor
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA.,Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - R Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - David J Lederer
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
23
|
Nishimura T, Ito S, Saito H, Hiver S, Shigetomi K, Ikenouchi J, Takeichi M. DAAM1 stabilizes epithelial junctions by restraining WAVE complex-dependent lateral membrane motility. J Cell Biol 2016; 215:559-573. [PMID: 27807130 PMCID: PMC5119936 DOI: 10.1083/jcb.201603107] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/13/2016] [Accepted: 10/14/2016] [Indexed: 12/31/2022] Open
Abstract
Nishimura et al. show that DAAM1, a formin family actin polymerization regulator, stabilizes epithelial cell junctions by counteracting the WAVE complex, another actin regulator. Loss of DAAM1 promotes the motility of junctional membranes and thereby enhances their invasion of neighboring environments. Epithelial junctions comprise two subdomains, the apical junctional complex (AJC) and the adjacent lateral membrane contacts (LCs), that span the majority of the junction. The AJC is lined with circumferential actin cables, whereas the LCs are associated with less-organized actin filaments whose roles are elusive. We found that DAAM1, a formin family actin regulator, accumulated at the LCs, and its depletion caused dispersion of actin filaments at these sites while hardly affecting circumferential actin cables. DAAM1 loss enhanced the motility of LC-forming membranes, leading to their invasion of neighboring cell layers, as well as disruption of polarized epithelial layers. We found that components of the WAVE complex and its downstream targets were required for the elevation of LC motility caused by DAAM1 loss. These findings suggest that the LC membranes are motile by nature because of the WAVE complex, but DAAM1-mediated actin regulation normally restrains this motility, thereby stabilizing epithelial architecture, and that DAAM1 loss evokes invasive abilities of epithelial cells.
Collapse
Affiliation(s)
- Tamako Nishimura
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Shoko Ito
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroko Saito
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Sylvain Hiver
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | - Kenta Shigetomi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | - Junichi Ikenouchi
- Department of Biology, Faculty of Sciences, Kyushu University, Nishi-Ku, Fukuoka 819-0395, Japan
| | | |
Collapse
|
24
|
Lian G, Dettenhofer M, Lu J, Downing M, Chenn A, Wong T, Sheen V. Filamin A- and formin 2-dependent endocytosis regulates proliferation via the canonical Wnt pathway. Development 2016; 143:4509-4520. [PMID: 27789627 DOI: 10.1242/dev.139295] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/18/2016] [Indexed: 01/07/2023]
Abstract
Actin-associated proteins regulate multiple cellular processes, including proliferation and differentiation, but the molecular mechanisms underlying these processes are unclear. Here, we report that the actin-binding protein filamin A (FlnA) physically interacts with the actin-nucleating protein formin 2 (Fmn2). Loss of FlnA and Fmn2 impairs proliferation, thereby generating multiple embryonic phenotypes, including microcephaly. FlnA interacts with the Wnt co-receptor Lrp6. Loss of FlnA and Fmn2 impairs Lrp6 endocytosis, downstream Gsk3β activity, and β-catenin accumulation in the nucleus. The proliferative defect in Flna and Fmn2 null neural progenitors is rescued by inhibiting Gsk3β activity. Our findings thus reveal a novel mechanism whereby actin-associated proteins regulate proliferation by mediating the endocytosis and transportation of components in the canonical Wnt pathway. Moreover, the Fmn2-dependent signaling in this pathway parallels that seen in the non-canonical Wnt-dependent regulation of planar cell polarity through the Formin homology protein Daam. These studies provide evidence for integration of actin-associated processes in directing neuroepithelial proliferation.
Collapse
Affiliation(s)
- Gewei Lian
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Markus Dettenhofer
- Central European Institute of Technology, Žerotínovo nám. 9, Brno 601 77, Czech Republic
| | - Jie Lu
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Michael Downing
- Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Anjen Chenn
- Department of Pathology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Timothy Wong
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Volney Sheen
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
25
|
Li J, Krishna SM, Golledge J. The Potential Role of Kallistatin in the Development of Abdominal Aortic Aneurysm. Int J Mol Sci 2016; 17:ijms17081312. [PMID: 27529213 PMCID: PMC5000709 DOI: 10.3390/ijms17081312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a vascular condition that causes permanent dilation of the abdominal aorta, which can lead to death due to aortic rupture. The only treatment for AAA is surgical repair, and there is no current drug treatment for AAA. Aortic inflammation, vascular smooth muscle cell apoptosis, angiogenesis, oxidative stress and vascular remodeling are implicated in AAA pathogenesis. Kallistatin is a serine proteinase inhibitor, which has been shown to have a variety of functions, potentially relevant in AAA pathogenesis. Kallistatin has been reported to have inhibitory effects on tumor necrosis factor alpha (TNF-α) signaling induced oxidative stress and apoptosis. Kallistatin also inhibits vascular endothelial growth factor (VEGF) and Wnt canonical signaling, which promote inflammation, angiogenesis, and vascular remodeling in various pre-clinical experimental models. This review explores the potential protective role of kallistatin in AAA pathogenesis.
Collapse
Affiliation(s)
- Jiaze Li
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, 4811 Townsville, Australia.
| | - Smriti Murali Krishna
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, 4811 Townsville, Australia.
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, 4811 Townsville, Australia.
- Department of Vascular and Endovascular Surgery, The Townsville Hospital, 4811 Townsville, Australia.
| |
Collapse
|
26
|
Zhan YH, Luo QC, Zhang XR, Xiao NA, Lu CX, Yue C, Wang N, Ma QL. CELSR1 is a positive regulator of endothelial cell migration and angiogenesis. BIOCHEMISTRY (MOSCOW) 2016; 81:591-9. [DOI: 10.1134/s0006297916060055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
27
|
Pariante P, Dotolo R, Venditti M, Ferrara D, Donizetti A, Aniello F, Minucci S. First Evidence of DAAM1 Localization During the Post-Natal Development of Rat Testis and in Mammalian Sperm. J Cell Physiol 2016; 231:2172-84. [PMID: 26831620 DOI: 10.1002/jcp.25330] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/01/2016] [Indexed: 01/16/2023]
Abstract
Dishevelled-associated activator of morphogenesis 1 (DAAM1) is a formin-family protein involved in nucleation of unbranched actin filaments and in cytoskeletal organization through Wnt-Dishevelled PCP pathway, which participates in essential biological processes, such as cell polarity, movement, and adhesion during morphogenesis and organogenesis. While its role has been investigated during development and in somatic cells, its potential association with the germinal compartment and reproduction is still unexplored. In this work, we assessed the possible association of DAAM1 with the morphogenesis of rat testis. We studied its expression and profiled its localization versus actin and tubulin, during the first wave of spermatogenesis and in the adult gonad (from 7 to 60 dpp). We show that, in mitotic phases, DAAM1 shares its localization with actin in Sertoli cells, gonocytes, and spermatogonia. Later, during meiosis, both proteins are found in spermatocytes, while only actin is detectable at the forming blood-testis barrier. DAAM1, then, follows the development of the acrosome system throughout spermiogenesis, and it is finally retained inside the cytoplasmic droplet in mature gametes, as corroborated by additional immunolocalization data on both rat and human sperm. Unlike the DAAM1, actin keeps its localization in Sertoli cells, and tubulin is associated with their protruding cytoplasm during the process. Our data support, for the first time, the hypothesis of a role for DAAM1 in cytoskeletal organization during Mammalian testis morphogenesis and gamete progression, while also hinting at its possible investigation as a morphological marker of germ cell and sperm physiology. J. Cell. Physiol. 231: 2172-2184, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paolo Pariante
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Seconda Università di Napoli, Napoli, Italy
| | - Raffaele Dotolo
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Seconda Università di Napoli, Napoli, Italy
| | - Massimo Venditti
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Seconda Università di Napoli, Napoli, Italy
| | - Diana Ferrara
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Seconda Università di Napoli, Napoli, Italy
| | - Aldo Donizetti
- Dipartimento di Biologia, Università di Napoli Federico II, Napoli, Italy
| | - Francesco Aniello
- Dipartimento di Biologia, Università di Napoli Federico II, Napoli, Italy
| | - Sergio Minucci
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate "F. Bottazzi", Seconda Università di Napoli, Napoli, Italy
| |
Collapse
|
28
|
Guillabert-Gourgues A, Jaspard-Vinassa B, Bats ML, Sewduth RN, Franzl N, Peghaire C, Jeanningros S, Moreau C, Roux E, Larrieu-Lahargue F, Dufourcq P, Couffinhal T, Duplàa C. Kif26b controls endothelial cell polarity through the Dishevelled/Daam1-dependent planar cell polarity-signaling pathway. Mol Biol Cell 2016; 27:941-53. [PMID: 26792835 PMCID: PMC4791138 DOI: 10.1091/mbc.e14-08-1332] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/13/2016] [Indexed: 11/11/2022] Open
Abstract
Angiogenesis involves the coordinated growth and migration of endothelial cells (ECs) toward a proangiogenic signal. The Wnt planar cell polarity (PCP) pathway, through the recruitment of Dishevelled (Dvl) and Dvl-associated activator of morphogenesis (Daam1), has been proposed to regulate cell actin cytoskeleton and microtubule (MT) reorganization for oriented cell migration. Here we report that Kif26b--a kinesin--and Daam1 cooperatively regulate initiation of EC sprouting and directional migration via MT reorganization. First, we find that Kif26b is recruited within the Dvl3/Daam1 complex. Using a three-dimensional in vitro angiogenesis assay, we show that Kif26b and Daam1 depletion impairs tip cell polarization and destabilizes extended vascular processes. Kif26b depletion specifically alters EC directional migration and mislocalized MT organizing center (MTOC)/Golgi and myosin IIB cell rear enrichment. Therefore the cell fails to establish a proper front-rear polarity. Of interest, Kif26b ectopic expression rescues the siDaam1 polarization defect phenotype. Finally, we show that Kif26b functions in MT stabilization, which is indispensable for asymmetrical cell structure reorganization. These data demonstrate that Kif26b, together with Dvl3/Daam1, initiates cell polarity through the control of PCP signaling pathway-dependent activation.
Collapse
Affiliation(s)
| | - Beatrice Jaspard-Vinassa
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | - Marie-Lise Bats
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | - Raj N Sewduth
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Nathalie Franzl
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Claire Peghaire
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Sylvie Jeanningros
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Catherine Moreau
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France
| | - Etienne Roux
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | | | - Pascale Dufourcq
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| | - Thierry Couffinhal
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France Service des Maladies Cardiaques et Vasculaires, Centre Hospitalier Universitaire de Bordeaux, F-33000 Bordeaux, France
| | - Cecile Duplàa
- Adaptation Cardiovasculaire à l'Ischémie, INSERM, U1034, F-33600 Pessac, France Adaptation Cardiovasculaire à l'Ischémie, U1034, Université de Bordeaux, F-33600 Pessac, France
| |
Collapse
|
29
|
Chen SY, Han B, Zhu YT, Mahabole M, Huang J, Beebe DC, Tseng SCG. HC-HA/PTX3 Purified From Amniotic Membrane Promotes BMP Signaling in Limbal Niche Cells to Maintain Quiescence of Limbal Epithelial Progenitor/Stem Cells. Stem Cells 2015; 33:3341-55. [PMID: 26148958 DOI: 10.1002/stem.2091] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/08/2015] [Accepted: 05/23/2015] [Indexed: 12/13/2022]
Abstract
To explore how limbal niche cells (LNCs) may control quiescence, self-renewal, and corneal epithelial lineage commitment/differentiation of limbal epithelial progenitor/stem cells (LEPCs), we have established an in vitro sphere assay by reunion between the two cell types in three-dimensional Matrigel. The resultant sphere exhibits inhibition of corneal epithelial lineage commitment/differentiation and marked clonal growth of LEPCs, of which the latter is correlated with activation of canonical Wnt signaling. Herein, we have created a similar reunion assay in immobilized heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3), which is purified from amniotic membrane (AM) and consists of a complex formed by hyaluronic covalently linked to heavy chain 1 of inter-α-inhibitor and noncovalently linked to pentraxin 3. The resultant spheres exhibited similar suppression of corneal epithelial lineage commitment/differentiation but upregulation of quiescence markers including nuclear translocation of Bmi-1, and negligible clonal growth of LEPCs. This outcome was correlated with the suppression of canonical Wnt but activation of noncanonical (Planar cell polarity) Wnt signaling as well as BMP signaling in both LEPCs and LNCs. The activation of BMP signaling in LNCs was pivotal because nuclear translocation of pSmad1/5/8 was prohibited in hLEPCs when reunioned with mLNCs of conditionally deleted Bmpr1a;Acvr1(DCKO) mice. Furthermore, ablation of BMP signaling in LEPCs led to upregulation of cell cycle genes, downregulation of Bmi-1, nuclear exclusion of phosphorylated Bmi-1, and marked promotion of the clonal growth of LEPCs. Hence, HC-HA/PTX3 uniquely upregulates BMP signaling in LNCs which leads to BMP signaling in LEPCs to achieve quiescence, helping explain how AM transplantation is clinically useful to be used as a matrix for ex vivo expansion of LEPCs and to treat corneal blindness caused by limbal stem cells deficiency.
Collapse
Affiliation(s)
- Szu-Yu Chen
- R&D Department, TissueTech, Inc., Miami, Florida, USA
| | - Bo Han
- Ocular Surface Research & Education Foundation, Miami, Florida, USA.,Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ying-Ting Zhu
- R&D Department, TissueTech, Inc., Miami, Florida, USA
| | | | - Jie Huang
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, USA
| | - David C Beebe
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, USA
| | - Scheffer C G Tseng
- R&D Department, TissueTech, Inc., Miami, Florida, USA.,Ocular Surface Research & Education Foundation, Miami, Florida, USA
| |
Collapse
|
30
|
Chapouly C, Tadesse Argaw A, Horng S, Castro K, Zhang J, Asp L, Loo H, Laitman BM, Mariani JN, Straus Farber R, Zaslavsky E, Nudelman G, Raine CS, John GR. Astrocytic TYMP and VEGFA drive blood-brain barrier opening in inflammatory central nervous system lesions. Brain 2015; 138:1548-67. [PMID: 25805644 DOI: 10.1093/brain/awv077] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/26/2015] [Indexed: 12/21/2022] Open
Abstract
In inflammatory central nervous system conditions such as multiple sclerosis, breakdown of the blood-brain barrier is a key event in lesion pathogenesis, predisposing to oedema, excitotoxicity, and ingress of plasma proteins and inflammatory cells. Recently, we showed that reactive astrocytes drive blood-brain barrier opening, via production of vascular endothelial growth factor A (VEGFA). Here, we now identify thymidine phosphorylase (TYMP; previously known as endothelial cell growth factor 1, ECGF1) as a second key astrocyte-derived permeability factor, which interacts with VEGFA to induce blood-brain barrier disruption. The two are co-induced NFκB1-dependently in human astrocytes by the cytokine interleukin 1 beta (IL1B), and inactivation of Vegfa in vivo potentiates TYMP induction. In human central nervous system microvascular endothelial cells, VEGFA and the TYMP product 2-deoxy-d-ribose cooperatively repress tight junction proteins, driving permeability. Notably, this response represents part of a wider pattern of endothelial plasticity: 2-deoxy-d-ribose and VEGFA produce transcriptional programs encompassing angiogenic and permeability genes, and together regulate a third unique cohort. Functionally, each promotes proliferation and viability, and they cooperatively drive motility and angiogenesis. Importantly, introduction of either into mouse cortex promotes blood-brain barrier breakdown, and together they induce severe barrier disruption. In the multiple sclerosis model experimental autoimmune encephalitis, TYMP and VEGFA co-localize to reactive astrocytes, and correlate with blood-brain barrier permeability. Critically, blockade of either reduces neurologic deficit, blood-brain barrier disruption and pathology, and inhibiting both in combination enhances tissue preservation. Suggesting importance in human disease, TYMP and VEGFA both localize to reactive astrocytes in multiple sclerosis lesion samples. Collectively, these data identify TYMP as an astrocyte-derived permeability factor, and suggest TYMP and VEGFA together promote blood-brain barrier breakdown.
Collapse
Affiliation(s)
- Candice Chapouly
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Azeb Tadesse Argaw
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Sam Horng
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Kamilah Castro
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jingya Zhang
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Linnea Asp
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Hannah Loo
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Benjamin M Laitman
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - John N Mariani
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Rebecca Straus Farber
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Elena Zaslavsky
- 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 4 Department of Systems Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - German Nudelman
- 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 4 Department of Systems Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Cedric S Raine
- 5 Department of Pathology (Neuropathology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gareth R John
- 1 Corinne Goldsmith Dickinson Centre for MS, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 2 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA 3 Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
31
|
Ossipova O, Chuykin I, Chu CW, Sokol SY. Vangl2 cooperates with Rab11 and Myosin V to regulate apical constriction during vertebrate gastrulation. Development 2014; 142:99-107. [PMID: 25480917 DOI: 10.1242/dev.111161] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Core planar cell polarity (PCP) proteins are well known to regulate polarity in Drosophila and vertebrate epithelia; however, their functions in vertebrate morphogenesis remain poorly understood. In this study, we describe a role for PCP signaling in the process of apical constriction during Xenopus gastrulation. The core PCP protein Vangl2 is detected at the apical surfaces of cells at the blastopore lip, and it functions during blastopore formation and closure. Further experiments show that Vangl2, as well as Daam1 and Rho-associated kinase (Rock), regulate apical constriction of bottle cells at the blastopore and ectopic constriction of ectoderm cells triggered by the actin-binding protein Shroom3. At the blastopore lip, Vangl2 is required for the apical accumulation of the recycling endosome marker Rab11. We also show that Rab11 and the associated motor protein Myosin V play essential roles in both endogenous and ectopic apical constriction, and might be involved in Vangl2 trafficking to the cell surface. Overexpression of Rab11 RNA was sufficient to partly restore normal blastopore formation in Vangl2-deficient embryos. These observations suggest that Vangl2 affects Rab11 to regulate apical constriction during blastopore formation.
Collapse
Affiliation(s)
- Olga Ossipova
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilya Chuykin
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chih-Wen Chu
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
32
|
Prokop A, Sánchez-Soriano N, Gonçalves-Pimentel C, Molnár I, Kalmár T, Mihály J. DAAM family members leading a novel path into formin research. Commun Integr Biol 2014. [DOI: 10.4161/cib.16511] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
33
|
Sewduth RN, Jaspard-Vinassa B, Peghaire C, Guillabert A, Franzl N, Larrieu-Lahargue F, Moreau C, Fruttiger M, Dufourcq P, Couffinhal T, Duplàa C. The ubiquitin ligase PDZRN3 is required for vascular morphogenesis through Wnt/planar cell polarity signalling. Nat Commun 2014; 5:4832. [PMID: 25198863 DOI: 10.1038/ncomms5832] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 07/28/2014] [Indexed: 01/01/2023] Open
Abstract
Development and stabilization of a vascular plexus requires the coordination of multiple signalling processes. Wnt planar cell polarity (PCP) signalling is critical in vertebrates for diverse morphogenesis events, which coordinate cell orientation within a tissue-specific plane. However, its functional role in vascular morphogenesis is not well understood. Here we identify PDZRN3, an ubiquitin ligase, and report that Pdzrn3 deficiency impairs embryonic angiogenic remodelling and postnatal retinal vascular patterning, with a loss of two-dimensional polarized orientation of the intermediate retinal plexus. Using in vitro and ex vivo Pdzrn3 loss-of-function and gain-of-function experiments, we demonstrate a key role of PDZRN3 in endothelial cell directional and coordinated extension. PDZRN3 ubiquitinates Dishevelled 3 (Dvl3), to promote endocytosis of the Frizzled/Dvl3 complex, for PCP signal transduction. These results highlight the role of PDZRN3 to direct Wnt PCP signalling, and broadly implicate this pathway in the planar orientation and highly branched organization of vascular plexuses.
Collapse
Affiliation(s)
- Raj N Sewduth
- INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | - Béatrice Jaspard-Vinassa
- 1] INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France [2] Univ. Bordeaux, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | - Claire Peghaire
- INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | - Aude Guillabert
- INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | - Nathalie Franzl
- INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | | | - Catherine Moreau
- INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | | | - Pascale Dufourcq
- 1] INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France [2] Univ. Bordeaux, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| | - Thierry Couffinhal
- 1] INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France [2] Univ. Bordeaux, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France [3] CHU de Bordeaux, Service des Maladies Cardiaques et Vasculaires, F-33000 Bordeaux, France
| | - Cécile Duplàa
- 1] INSERM, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France [2] Univ. Bordeaux, Adaptation cardiovasculaire à l'ischémie, U1034, F-33600 Pessac, France
| |
Collapse
|
34
|
Zhan YH, Lin Y, Tong SJ, Ma QL, Lu CX, Fang L, Wei W, Cai B, Wang N. The CELSR1 polymorphisms rs6007897 and rs4044210 are associated with ischaemic stroke in Chinese Han population. Ann Hum Biol 2014; 42:26-30. [PMID: 25117632 DOI: 10.3109/03014460.2014.944214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Recently, CELSR1 was identified by genome-wide association studies (GWAS) as a susceptibility gene for ischaemic stroke (IS) in Japanese individuals. AIM The goal was to examine whether CELSR1 variants are associated with IS in the Chinese Han population. SUBJECTS AND METHODS This study genotyped two single nucleotide polymorphisms (SNPs) of CELSR1, rs6007897 and rs4044210, in a Chinese sample of 569 IS cases and 581 controls and assessed their genotype and allele associations with IS. RESULTS The results showed that rs6007897 and rs4044210 variants of CELSR1 were significantly (p < 0.01) associated with IS. These associations remained after adjustment for age, gender, smoking status, hypertension, diabetes mellitus and hypercholesterolemia. In addition, a significant association was observed of rs6007897 and rs4044210 of CELSR1 with large artery atherosclerosis (LAA), a sub-type of IS (p < 0.01). CONCLUSION Taken together, the present study has proven for the first time that CELSR1 is a susceptibility gene for IS in the Chinese Han population, especially for LAA.
Collapse
Affiliation(s)
- Yi-Hong Zhan
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Center of Neuroscience, Fujian Medical University , Fuzhou , PR China and
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Paul I, Bhattacharya S, Chatterjee A, Ghosh MK. Current Understanding on EGFR and Wnt/β-Catenin Signaling in Glioma and Their Possible Crosstalk. Genes Cancer 2014; 4:427-46. [PMID: 24386505 DOI: 10.1177/1947601913503341] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/31/2013] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiformes (GBMs) are extensively heterogeneous at both cellular and molecular levels. Current therapeutic strategies include targeting of key signaling molecules using pharmacological inhibitors in combination with genotoxic agents such as temozolomide. In spite of all efforts, the prognosis of glioma patients remains dismal. Therefore, a proper understanding of individual molecular pathways responsible for the progression of GBM is necessary. The epidermal growth factor receptor (EGFR) pathway is probably the most significant signaling pathway clinically implicated in glioma. Not surprisingly, anti-EGFR therapies mostly prevail for therapeutic purposes. The Wnt/β-catenin pathway is well implicated in multiple tumors; however, its role in glioma has only recently started to emerge. We give a concise account of the current understanding of the role of both these pathways in glioma. Last, taking evidences from a limited literature, we outline a number of points where these pathways intersect each other and put forward the possibility of combinatorially targeting them for treatment of glioma.
Collapse
Affiliation(s)
- Indranil Paul
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Seemana Bhattacharya
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Anirban Chatterjee
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mrinal K Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
36
|
Hoffmann AK, Naj X, Linder S. Daam1 is a regulator of filopodia formation and phagocytic uptake of Borrelia burgdorferi by primary human macrophages. FASEB J 2014; 28:3075-89. [PMID: 24696301 DOI: 10.1096/fj.13-247049] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease, an infectious disease that primarily affects the skin, nervous system, and joints. Uptake of borreliae by immune cells is decisive for the course of the infection, and remodelling of the host actin cytoskeleton is crucial in this process. In this study, we showed that the actin-regulatory formin Daam1 is important in Borrelia phagocytosis by primary human macrophages. Uptake of borreliae proceeds preferentially through capture by filopodia and formation of coiling pseudopods that enwrap the spirochetes. Using immunofluorescence, we localized endogenous and overexpressed Daam1 to filopodia and to F-actin-rich uptake structures. Live-cell imaging further showed that Daam1 is enriched at coiling pseudopods that arise from the macrophage surface. This filopodia-independent step was corroborated by control experiments of phagocytic cup formation with latex beads. Moreover, siRNA-mediated knockdown of Daam1 led to a 65% reduction of borreliae-induced filopodia, and, as shown by the outside-inside staining technique, to a 50% decrease in phagocytic uptake of borreliae, as well as a 37% reduction in coiling pseudopod formation. Collectively, we showed that Daam1 plays a dual role in the phagocytic uptake of borreliae: first, as a regulator of filopodia, which are used for capturing spirochetes, and second, in the formation of the coiling pseudopod that enwraps the bacterial cell. These data identify Daam1 as a novel regulator of B. burgdorferi phagocytosis. At the same time, this is the first demonstration of a role for Daam1 in phagocytic processes in general.-Hoffmann, A.-K., Naj, X., Linder, S. Daam1 is a regulator of filopodia formation and phagocytic uptake of Borrelia burgdorferi by primary human macrophages.
Collapse
Affiliation(s)
- Ann-Kathrin Hoffmann
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Xenia Naj
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
37
|
The Wnt/planar cell polarity signaling pathway contributes to the integrity of tight junctions in brain endothelial cells. J Cereb Blood Flow Metab 2014; 34:433-40. [PMID: 24346691 PMCID: PMC3948118 DOI: 10.1038/jcbfm.2013.213] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/14/2013] [Accepted: 11/07/2013] [Indexed: 11/08/2022]
Abstract
Wnt morphogens released by neural precursor cells were recently reported to control blood-brain barrier (BBB) formation during development. Indeed, in mouse brain endothelial cells, activation of the Wnt/β-catenin signaling pathway, also known as the canonical Wnt pathway, was shown to stabilize endothelial tight junctions (TJs) through transcriptional regulation of the expression of TJ proteins. Because Wnt proteins activate several distinct β-catenin-dependent and independent signaling pathways, this study was designed to assess whether the noncanonical Wnt/Par/aPKC planar cell polarity (PCP) pathway might also control TJ integrity in brain endothelial cells. First we established, in the hCMEC/D3 human brain endothelial cell line, that the Par/aPKC PCP complex colocalizes with TJs and controls apicobasal polarization. Second, using an siRNA approach, we showed that the Par/aPKC PCP complex regulates TJ stability and reassembling after osmotic shock. Finally, we provided evidence that Wnt5a signals in hCMEC/D3 cells through activation of the Par/aPKC PCP complex, independently of the Wnt canonical β-catenin-dependent pathway and significantly contributes to TJ integrity and endothelial apicobasal polarity. In conclusion, this study suggests that the Wnt/Par/aPKC PCP pathway, in addition to the Wnt/β-catenin canonical pathway, is a key regulator of the BBB.
Collapse
|
38
|
Paz AC, Soleas J, Poon JC, Trieu D, Waddell TK, McGuigan AP. Challenges and Opportunities for Tissue-Engineering Polarized Epithelium. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:56-72. [DOI: 10.1089/ten.teb.2013.0144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ana C. Paz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - John Soleas
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - James C.H. Poon
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Dennis Trieu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Thomas K. Waddell
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
WNT signaling in neoplasia. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
40
|
Colombo A, Palma K, Armijo L, Mione M, Signore IA, Morales C, Guerrero N, Meynard MM, Pérez R, Suazo J, Marcelain K, Briones L, Härtel S, Wilson SW, Concha ML. Daam1a mediates asymmetric habenular morphogenesis by regulating dendritic and axonal outgrowth. Development 2013; 140:3997-4007. [PMID: 24046318 PMCID: PMC3775416 DOI: 10.1242/dev.091934] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Although progress has been made in resolving the genetic pathways that specify neuronal asymmetries in the brain, little is known about genes that mediate the development of structural asymmetries between neurons on left and right. In this study, we identify daam1a as an asymmetric component of the signalling pathways leading to asymmetric morphogenesis of the habenulae in zebrafish. Daam1a is a member of the Formin family of actin-binding proteins and the extent of Daam1a expression in habenular neuron dendrites mirrors the asymmetric growth of habenular neuropil between left and right. Local loss and gain of Daam1a function affects neither cell number nor subtype organisation but leads to a decrease or increase of neuropil, respectively. Daam1a therefore plays a key role in the asymmetric growth of habenular neuropil downstream of the pathways that specify asymmetric cellular domains in the habenulae. In addition, Daam1a mediates the development of habenular efferent connectivity as local loss and gain of Daam1a function impairs or enhances, respectively, the growth of habenular neuron terminals in the interpeduncular nucleus. Abrogation of Daam1a disrupts the growth of both dendritic and axonal processes and results in disorganised filamentous actin and α-tubulin. Our results indicate that Daam1a plays a key role in asymmetric habenular morphogenesis mediating the growth of dendritic and axonal processes in dorsal habenular neurons.
Collapse
Affiliation(s)
- Alicia Colombo
- Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Reis M, Liebner S. Wnt signaling in the vasculature. Exp Cell Res 2013; 319:1317-23. [PMID: 23291327 DOI: 10.1016/j.yexcr.2012.12.023] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 12/21/2022]
Abstract
The development of the vascular system requires orchestrated activities of various molecular pathways to assure the formation of a hierarchically branched tubular network. Furthermore, endothelial cell (EC) populations are heterogeneous to meet organ-specific requirements in the mature vasculature. This developmental scheme is probably best represented by the acquisition and maintenance of unique barrier properties known as the blood-brain barrier (BBB) in microvessels of the central nervous system (CNS). Only recently, the canonical Wnt/β-catenin pathway was implicated in many aspects of angiogenesis, vascular remodeling and differentiation in various species and organ systems. Beside its major contribution to brain angiogenesis and barrier formation, the Wnt/β-catenin pathway influences vascular sprouting, remodeling and arterio-venous specification by modulating the Notch pathway. Furthermore, canonical Wnt signaling has been implicated in heart valve formation by initiating endothelial-mesenchymal transition. Growing evidence also points to a role of the non-canonical Wnt pathway in vascular development by regulating VEGF availability. Several novel findings regarding the role of the Wnt pathway in developmental as well as in pathological angiogenesis prompted us to review its emerging function in the vasculature.
Collapse
Affiliation(s)
- Marco Reis
- Institute of Neurology (Edinger-Institute), Johann Wolfgang Goethe-University Frankfurt Medical School, Heinrich-Hoffmann-Straße 7, 60528 Frankfurt, Germany
| | | |
Collapse
|
42
|
Choi HJ, Park H, Lee HW, Kwon YG. The Wnt pathway and the roles for its antagonists, DKKS, in angiogenesis. IUBMB Life 2012; 64:724-31. [PMID: 22807036 DOI: 10.1002/iub.1062] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/06/2012] [Accepted: 05/24/2012] [Indexed: 11/07/2022]
Abstract
The Wnt signaling pathway is involved in a wide range of developmental and physiological processes, such as cell fate specification, tissue morphogenesis, and homeostasis. Thus, its dysregulation has been found in multiple diseases, including some cardiovascular disorders. The loss or gain of function of Wnt pathway components results in abnormal vascular development and angiogenesis. Further study has revealed that Wnt signaling in endothelial cells appears to contribute to vascular morphogenesis and endothelial cell specification. Owing to the significance of Wnt signaling in angiogenesis, Wnt antagonists have been considered potential treatments for neovascular disorders. In line with this, members of the Dkk protein family (Dkks), well-known Wnt antagonists, have been recently found to regulate angiogenesis. This review summarizes our present knowledge of the roles of Wnt signaling and Wnt antagonists, particularly Dkks, in angiogenic regulation and explores the therapeutic potential of Wnt antagonists.
Collapse
Affiliation(s)
- Hyun-Jung Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
43
|
Sundberg TB, Darricarrere N, Cirone P, Li X, McDonald L, Mei X, Westlake CJ, Slusarski DC, Beynon RJ, Crews CM. Disruption of Wnt planar cell polarity signaling by aberrant accumulation of the MetAP-2 substrate Rab37. ACTA ACUST UNITED AC 2012; 18:1300-11. [PMID: 22035799 DOI: 10.1016/j.chembiol.2011.07.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 07/18/2011] [Accepted: 07/28/2011] [Indexed: 11/25/2022]
Abstract
Identification of methionine aminopeptidase-2 (MetAP-2) as the molecular target of the antiangiogenic compound TNP-470 has sparked interest in N-terminal Met excision's (NME) role in endothelial cell biology. In this regard, we recently demonstrated that MetAP-2 inhibition suppresses Wnt planar cell polarity (PCP) signaling and that endothelial cells depend on this pathway for normal function. Despite this advance, the substrate(s) whose activity is altered upon MetAP-2 inhibition, resulting in loss of Wnt PCP signaling, is not known. Here we identify the small G protein Rab37 as a MetAP-2-specific substrate that accumulates in the presence of TNP-470. A functional role for aberrant Rab37 accumulation in TNP-470's mode of action is demonstrated using a Rab37 point mutant that is resistant to NME, because expression of this mutant phenocopies the effects of MetAP-2 inhibition on Wnt PCP signaling-dependent processes.
Collapse
Affiliation(s)
- Thomas B Sundberg
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Beane WS, Tseng AS, Morokuma J, Lemire JM, Levin M. Inhibition of planar cell polarity extends neural growth during regeneration, homeostasis, and development. Stem Cells Dev 2012; 21:2085-94. [PMID: 22339734 DOI: 10.1089/scd.2011.0605] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The ability to stop producing or replacing cells at the appropriate time is essential, as uncontrolled growth can lead to loss of function and even cancer. Tightly regulated mechanisms coordinate the growth of stem cell progeny with the patterning needs of the host organism. Despite the importance of proper termination during regeneration, cell turnover, and embryonic development, very little is known about how tissues determine when patterning is complete during these processes. Using planarian flatworms, we show that the planar cell polarity (PCP) pathway is required to stop the growth of neural tissue. Although traditionally studied as regulators of tissue polarity, we found that loss of the PCP genes Vangl2, DAAM1, and ROCK by RNA interference (individually or together) resulted in supernumerary eyes and excess optical neurons in intact planarians, while regenerating planarians had continued hyperplasia throughout the nervous system long after controls ceased new growth. This failure to terminate growth suggests that neural tissues use PCP as a readout of patterning, highlighting a potential role for intact PCP as a signal to stem and progenitor cells to halt neuronal growth when patterning is finished. Moreover, we found this mechanism to be conserved in vertebrates. Loss of Vangl2 during normal development, as well as during Xenopus tadpole tail regeneration, also leads to the production of excess neural tissue. This evolutionarily conserved function of PCP represents a tractable new approach for controlling the growth of nerves.
Collapse
Affiliation(s)
- Wendy S Beane
- Biology Department and Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
In mammals, the skin can form complex global and local patterns to meet diverse functional requirements in different parts of the body. To date, the fundamental principles that underlie skin patterning remain poorly understood because of the involvement of multiple interacting processes. Genes involved in the planar cell polarity (PCP) signalling pathway, which is capable of polarizing cells within the planar plane of an epithelium, can control the orientation and differentiation of hair follicles, underlining their involvement in skin pattern formation. Here, we summarize recent progress that has been made to understand the PCP signalling pathway and its function in mammalian skin, including its role in hair follicle morphogenesis, ciliogenesis and wound healing. We argue that dissecting PCP signalling in the context of hair follicle formation might reveal many as-yet-undiscovered functions for PCP in the development, homeostasis and regeneration of skin.
Collapse
Affiliation(s)
- Jiang Chen
- Department of Dermatology, Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | |
Collapse
|
46
|
Descamps B, Sewduth R, Ferreira Tojais N, Jaspard B, Reynaud A, Sohet F, Lacolley P, Allières C, Lamazière JMD, Moreau C, Dufourcq P, Couffinhal T, Duplàa C. Frizzled 4 Regulates Arterial Network Organization Through Noncanonical Wnt/Planar Cell Polarity Signaling. Circ Res 2012; 110:47-58. [DOI: 10.1161/circresaha.111.250936] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Rationale:
A growing body of evidence supports the hypothesis that the Wnt/planar cell polarity (PCP) pathway regulates endothelial cell proliferation and angiogenesis, but the components that mediate this regulation remain elusive.
Objective:
We investigated the involvement of one of the receptors, Frizzled4 (Fzd4), in this process because its role has been implicated in retinal vascular development.
Methods and Results:
We found that loss of
fzd4
function in mice results in a striking reduction and impairment of the distal small artery network in the heart and kidney. We report that loss of
fzd4
decreases vascular cell proliferation and migration and decreases the ability of the endothelial cells to form tubes. We show that
fzd4
deletion induces defects in the expression level of stable acetylated tubulin and in Golgi organization during migration. Deletion of
fzd4
favors Wnt noncanonical AP1-dependent signaling, indicating that Fzd4 plays a pivotal role favoring PCP signaling. Our data further demonstrate that Fzd4 is predominantly localized on the top of the plasma membrane, where it preferentially induces Dvl3 relocalization to promote its activation and α-tubulin recruitment during migration. In a pathological mouse angiogenic model, deletion of
fzd4
impairs the angiogenic response and leads to the formation of a disorganized arterial network.
Conclusions:
These results suggest that Fzd4 is a major receptor involved in arterial formation and organization through a Wnt/PCP pathway.
Collapse
Affiliation(s)
- Betty Descamps
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Raj Sewduth
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Nancy Ferreira Tojais
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Béatrice Jaspard
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Annabel Reynaud
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Fabien Sohet
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Patrick Lacolley
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Cécile Allières
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Jean-Marie Daniel Lamazière
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Catherine Moreau
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Pascale Dufourcq
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Thierry Couffinhal
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| | - Cécile Duplàa
- From the Inserm (B.D., R.S., N.F.T., B.J., A.R., C.A., J.-M.D.L., C.M., P.D., T.C., C.D.), U1034, Pessac, France and University de Bordeaux, Bordeaux, France; Unité Inserm U872 (F.S.), Centre de Recherche des Cordeliers, Paris, France; Université Henri Poincaré (P.L., P.D.), Inserm U961, Nancy, France; Laboratoire de biochimie (T.C.), UFR Sciences Pharmaceutiques, University de Bordeaux, Bordeaux, France; Department of Cardiology (C.D.), Pôle Cardiothoracique, Hôpital Haut Lévêque, Pessac, France
| |
Collapse
|
47
|
Planutiene M, Planutis K, Holcombe RF. Lymphoid enhancer-binding factor 1, a representative of vertebrate-specific Lef1/Tcf1 sub-family, is a Wnt-beta-catenin pathway target gene in human endothelial cells which regulates matrix metalloproteinase-2 expression and promotes endothelial cell invasion. Vasc Cell 2011; 3:28. [PMID: 22168911 PMCID: PMC3269378 DOI: 10.1186/2045-824x-3-28] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 12/14/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Wnt signaling is activated in many types of cancer and normal physiological processes. Various Wnt-related secreted factors may influence angiogenesis both in the tumor microenvironment and in normal tissues by direct action on endothelial cells. The mechanism of this Wnt action in angiogenesis is not well defined. We hypothesize that endothelial cells are responsive to Wnt signals and that Lef1, a member of the vertebrate-specific Wnt/beta-catenin throughput-inducing transcription factors' sub-family Lef1/Tcf1, mediates this responsiveness and promotes endothelial cell invasion. METHODS A human endothelial cell line, EAhy926 was exposed to Wnt3a or directly transfected with Lef1. Readouts included assessment of nuclear beta-catenin, Wnt throughput with a SuperTOPflash reporter assay, induction of Lef1 transcription, induction of matrix metalloproteinase (MMP)-2 transcription, cell proliferation and cell invasion through a matrix in vitro. The effects on MMP2 were also evaluated in the presence of Lef1 silencing siRNA. RESULTS Wnt3a increased nuclear beta-catenin and up-regulated Wnt/beta-catenin throughput. Wnt3a increased Lef1 transcription and activity of the Lef1 promoter. Both Wnt3a treatment and Lef1 overexpression induced MMP2 transcription but this effect was completely abrogated in the presence of Lef1 siRNA. Inhibition of Lef1 also reduced basal MMP2 levels suggesting that Lef1 regulates MMP2 expression even in the absence of exogenous Wnt pathway activation. Lef1 slightly increased proliferation of EAhy926 cells and increased invasion by more than two-fold. CONCLUSIONS EAhy926 cells activate canonical Wnt signaling in response to Wnt3a ligand. The Wnt target Lef1 specifically regulates MMP2 expression in these cells and promotes endothelial cell invasion. The EAhy926 cell line provides a convenient alternative to primary human umbilical vein endothelial cells (HUVEC) in the study of angiogenesis and the role of Wnt signaling on endothelial cell function.
Collapse
Affiliation(s)
- Marina Planutiene
- The Tisch Cancer Institute at The Mount Sinai Medical Center, One Gustave L Levy Place, New York, NY 10029-6500, USA
| | - Kestutis Planutis
- The Tisch Cancer Institute at The Mount Sinai Medical Center, One Gustave L Levy Place, New York, NY 10029-6500, USA
| | - Randall F Holcombe
- The Tisch Cancer Institute at The Mount Sinai Medical Center, One Gustave L Levy Place, New York, NY 10029-6500, USA
| |
Collapse
|
48
|
Augustin I, Goidts V, Bongers A, Kerr G, Vollert G, Radlwimmer B, Hartmann C, Herold-Mende C, Reifenberger G, von Deimling A, Boutros M. The Wnt secretion protein Evi/Gpr177 promotes glioma tumourigenesis. EMBO Mol Med 2011; 4:38-51. [PMID: 22147553 PMCID: PMC3306557 DOI: 10.1002/emmm.201100186] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/16/2011] [Accepted: 10/21/2011] [Indexed: 11/08/2022] Open
Abstract
Malignant astrocytomas are highly aggressive brain tumours with poor prognosis. While a number of structural genomic changes and dysregulation of signalling pathways in gliomas have been described, the identification of biomarkers and druggable targets remains an important task for novel diagnostic and therapeutic approaches. Here, we show that the Wnt-specific secretory protein Evi (also known as GPR177/Wntless/Sprinter) is overexpressed in astrocytic gliomas. Evi/Wls is a core Wnt signalling component and a specific regulator of pan-Wnt protein secretion, affecting both canonical and non-canonical signalling. We demonstrate that its depletion in glioma and glioma-derived stem-like cells led to decreased cell proliferation and apoptosis. Furthermore, Evi/Wls silencing in glioma cells reduced cell migration and the capacity to form tumours in vivo. We further show that Evi/Wls overexpression is sufficient to promote downstream Wnt signalling. Taken together, our study identifies Evi/Wls as an essential regulator of glioma tumourigenesis, identifying a pathway-specific protein trafficking factor as an oncogene and offering novel therapeutic options to interfere with the aberrant regulation of growth factors at the site of production.
Collapse
Affiliation(s)
- Iris Augustin
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics and Heidelberg University, Faculty of Medicine Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zou Y. When it is hard to get to with genetics--planar cell polarity under a chemical scalpel. ACTA ACUST UNITED AC 2011; 18:1350-1. [PMID: 22118667 DOI: 10.1016/j.chembiol.2011.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Planar cell polarity (PCP) has been under genetic dissection for decades. More and more fundamental developmental processes have been found relying on PCP signaling. However, mechanisms of how PCP signaling generates asymmetry is still unknown. A recent paper in Chemistry & Biology (Sundberg et al., 2011) represents the efforts to decipher the intracellular code of polarity signaling.
Collapse
Affiliation(s)
- Yimin Zou
- Neurobiology Section. Biological Sciences Division. University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
50
|
Endothelial development taking shape. Curr Opin Cell Biol 2011; 23:676-85. [PMID: 22051380 DOI: 10.1016/j.ceb.2011.10.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 10/12/2011] [Indexed: 11/22/2022]
Abstract
Blood vessel development is a vital process during embryonic development, during tissue growth, regeneration and disease processes in the adult. In the past decade researchers have begun to unravel basic molecular mechanisms that regulate the formation of vascular lumen, sprouting angiogenesis, fusion of vessels, and pruning of the vascular plexus. The understanding of the biology of these angiogenic processes is increasingly driven through studies on vascular development at the cellular resolution. Single cell analysis in vivo, advanced genetic tools and the widespread use of powerful animal models combined with improved imaging possibilities are delivering new insights into endothelial cell form, function and behavior angiogenesis. Moreover, the combination of in silico modeling and experimentation including dynamic imaging promotes insights into higher level cooperative behavior leading to functional patterning of vascular networks. Here we summarize recent concepts and advances in the field of vascular development, focusing in detail on the endothelial cell.
Collapse
|