51
|
Nikolopoulou E, Galea GL, Rolo A, Greene NDE, Copp AJ. Neural tube closure: cellular, molecular and biomechanical mechanisms. Development 2017; 144:552-566. [PMID: 28196803 DOI: 10.1242/dev.145904] [Citation(s) in RCA: 341] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neural tube closure has been studied for many decades, across a range of vertebrates, as a paradigm of embryonic morphogenesis. Neurulation is of particular interest in view of the severe congenital malformations - 'neural tube defects' - that result when closure fails. The process of neural tube closure is complex and involves cellular events such as convergent extension, apical constriction and interkinetic nuclear migration, as well as precise molecular control via the non-canonical Wnt/planar cell polarity pathway, Shh/BMP signalling, and the transcription factors Grhl2/3, Pax3, Cdx2 and Zic2. More recently, biomechanical inputs into neural tube morphogenesis have also been identified. Here, we review these cellular, molecular and biomechanical mechanisms involved in neural tube closure, based on studies of various vertebrate species, focusing on the most recent advances in the field.
Collapse
Affiliation(s)
- Evanthia Nikolopoulou
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Gabriel L Galea
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Ana Rolo
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
52
|
Tseng CC, Jia B, Barndt R, Gu Y, Chen CY, Tseng IC, Su SF, Wang JK, Johnson MD, Lin CY. Matriptase shedding is closely coupled with matriptase zymogen activation and requires de novo proteolytic cleavage likely involving its own activity. PLoS One 2017; 12:e0183507. [PMID: 28829816 PMCID: PMC5567652 DOI: 10.1371/journal.pone.0183507] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/04/2017] [Indexed: 11/18/2022] Open
Abstract
The type 2 transmembrane serine protease matriptase is involved in many pathophysiological processes probably via its enzymatic activity, which depends on the dynamic relationship between zymogen activation and protease inhibition. Matriptase shedding can prolong the life of enzymatically active matriptase and increase accessibility to substrates. We show here that matriptase shedding occurs via a de novo proteolytic cleavage at sites located between the SEA domain and the CUB domain. Point or combined mutations at the four positively charged amino acid residues in the region following the SEA domain allowed Arg-186 to be identified as the primary cleavage site responsible for matriptase shedding. Kinetic studies further demonstrate that matriptase shedding is temporally coupled with matriptase zymogen activation. The onset of matriptase shedding lags one minute behind matriptase zymogen activation. Studies with active site triad Ser-805 point mutated matriptase, which no longer undergoes zymogen activation or shedding, further suggests that matriptase shedding depends on matriptase zymogen activation, and that matriptase proteolytic activity may be involved in its own shedding. Our studies uncover an autonomous mechanism coupling matriptase zymogen activation, proteolytic activity, and shedding such that a proportion of newly generated active matriptase escapes HAI-1-mediated rapid inhibition by shedding into the extracellular milieu.
Collapse
Affiliation(s)
- Chun-Che Tseng
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Bailing Jia
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
- Department of Gastroenterology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Robert Barndt
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Yayun Gu
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Chien-Yu Chen
- Department of Biochemistry National Defense Medical Center, Taipei, Taiwan
- School of Medicine National Defense Medical Center, Taipei, Taiwan
| | - I-Chu Tseng
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Sheng-Fang Su
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
| | - Jehng-Kang Wang
- Department of Biochemistry National Defense Medical Center, Taipei, Taiwan
| | - Michael D. Johnson
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
- * E-mail: (CYL); (MDJ)
| | - Chen-Yong Lin
- Lombardi Comprehensive Cancer Center, Department of Oncology Georgetown University, Washington DC, United States of America
- * E-mail: (CYL); (MDJ)
| |
Collapse
|
53
|
Isermann B. Homeostatic effects of coagulation protease-dependent signaling and protease activated receptors. J Thromb Haemost 2017; 15:1273-1284. [PMID: 28671351 DOI: 10.1111/jth.13721] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A homeostatic function of the coagulation system in regard to hemostasis is well established. Homeostasis of blood coagulation depends partially on protease activated receptor (PAR)-signaling. Beyond coagulation proteases, numerous other soluble and cell-bound proteases convey cellular effects via PAR signaling. As we learn more about the mechanisms underlying cell-, tissue-, and context-specific PAR signaling, we concurrently gain new insights into physiological and pathophysiological functions of PARs. In this regard, regulation of cell and tissue homeostasis by PAR signaling is an evolving scheme. Akin to the control of blood clotting per se (the fibrin-platelet interaction) coagulation proteases coordinately regulate cell- and tissue-specific functions. This review summarizes recent insights into homeostatic regulation through PAR signaling, focusing on blood coagulation proteases. Considering the common use of drugs altering coagulation protease activity through either broad or targeted inhibitory activities, and the advent of PAR modulating drugs, an in-depth understanding of the mechanisms through which coagulation proteases and PAR signaling regulate not only hemostasis, but also cell and tissue homeostasis is required.
Collapse
Affiliation(s)
- B Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
54
|
Galea GL, Cho YJ, Galea G, Molè MA, Rolo A, Savery D, Moulding D, Culshaw LH, Nikolopoulou E, Greene NDE, Copp AJ. Biomechanical coupling facilitates spinal neural tube closure in mouse embryos. Proc Natl Acad Sci U S A 2017; 114:E5177-E5186. [PMID: 28607062 PMCID: PMC5495245 DOI: 10.1073/pnas.1700934114] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neural tube (NT) formation in the spinal region of the mammalian embryo involves a wave of "zippering" that passes down the elongating spinal axis, uniting the neural fold tips in the dorsal midline. Failure of this closure process leads to open spina bifida, a common cause of severe neurologic disability in humans. Here, we combined a tissue-level strain-mapping workflow with laser ablation of live-imaged mouse embryos to investigate the biomechanics of mammalian spinal closure. Ablation of the zippering point at the embryonic dorsal midline causes far-reaching, rapid separation of the elevating neural folds. Strain analysis revealed tissue expansion around the zippering point after ablation, but predominant tissue constriction in the caudal and ventral neural plate zone. This zone is biomechanically coupled to the zippering point by a supracellular F-actin network, which includes an actin cable running along the neural fold tips. Pharmacologic inhibition of F-actin or laser ablation of the cable causes neural fold separation. At the most advanced somite stages, when completion of spinal closure is imminent, the cable forms a continuous ring around the neuropore, and simultaneously, a new caudal-to-rostral zippering point arises. Laser ablation of this new closure initiation point causes neural fold separation, demonstrating its biomechanical activity. Failure of spinal closure in pre-spina bifida Zic2Ku mutant embryos is associated with altered tissue biomechanics, as indicated by greater neuropore widening after ablation. Thus, this study identifies biomechanical coupling of the entire region of active spinal neurulation in the mouse embryo as a prerequisite for successful NT closure.
Collapse
Affiliation(s)
- Gabriel L Galea
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom;
| | - Young-June Cho
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Gauden Galea
- Division of Noncommunicable Diseases and Promoting Health Through the Life Course, World Health Organization Regional Office for Europe, Copenhagen DK-2100, Denmark
| | - Matteo A Molè
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Ana Rolo
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Dawn Savery
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Dale Moulding
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Lucy H Culshaw
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Evanthia Nikolopoulou
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| |
Collapse
|
55
|
Friis S, Tadeo D, Le-Gall SM, Jürgensen HJ, Sales KU, Camerer E, Bugge TH. Matriptase zymogen supports epithelial development, homeostasis and regeneration. BMC Biol 2017; 15:46. [PMID: 28571576 PMCID: PMC5452369 DOI: 10.1186/s12915-017-0384-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/05/2017] [Indexed: 12/31/2022] Open
Abstract
Background Matriptase is a membrane serine protease essential for epithelial development, homeostasis, and regeneration, as well as a central orchestrator of pathogenic pericellular signaling in the context of inflammatory and proliferative diseases. Matriptase is an unusual protease in that its zymogen displays measurable enzymatic activity. Results Here, we used gain and loss of function genetics to investigate the possible biological functions of zymogen matriptase. Unexpectedly, transgenic mice mis-expressing a zymogen-locked version of matriptase in the epidermis displayed pathologies previously reported for transgenic mice mis-expressing wildtype epidermal matriptase. Equally surprising, mice engineered to express only zymogen-locked endogenous matriptase, unlike matriptase null mice, were viable, developed epithelial barrier function, and regenerated the injured epithelium. Compatible with these observations, wildtype and zymogen-locked matriptase were equipotent activators of PAR-2 inflammatory signaling. Conclusion The study demonstrates that the matriptase zymogen is biologically active and is capable of executing developmental and homeostatic functions of the protease. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0384-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stine Friis
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.,Section for Molecular Disease Biology, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Tadeo
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.,Georgetown University School of Medicine, Washington, DC, 20057, USA
| | - Sylvain M Le-Gall
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Université Sorbonne Paris Cité, Paris, France
| | - Henrik Jessen Jürgensen
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA
| | - Katiuchia Uzzun Sales
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.,Department of Cell and Molecular Biology, Ribierão Preto School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France.,Université Sorbonne Paris Cité, Paris, France
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 320, Bethesda, MD, 20892, USA.
| |
Collapse
|
56
|
Mohd-Zin SW, Marwan AI, Abou Chaar MK, Ahmad-Annuar A, Abdul-Aziz NM. Spina Bifida: Pathogenesis, Mechanisms, and Genes in Mice and Humans. SCIENTIFICA 2017; 2017:5364827. [PMID: 28286691 PMCID: PMC5327787 DOI: 10.1155/2017/5364827] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/14/2016] [Accepted: 12/01/2016] [Indexed: 05/26/2023]
Abstract
Spina bifida is among the phenotypes of the larger condition known as neural tube defects (NTDs). It is the most common central nervous system malformation compatible with life and the second leading cause of birth defects after congenital heart defects. In this review paper, we define spina bifida and discuss the phenotypes seen in humans as described by both surgeons and embryologists in order to compare and ultimately contrast it to the leading animal model, the mouse. Our understanding of spina bifida is currently limited to the observations we make in mouse models, which reflect complete or targeted knockouts of genes, which perturb the whole gene(s) without taking into account the issue of haploinsufficiency, which is most prominent in the human spina bifida condition. We thus conclude that the need to study spina bifida in all its forms, both aperta and occulta, is more indicative of the spina bifida in surviving humans and that the measure of deterioration arising from caudal neural tube defects, more commonly known as spina bifida, must be determined by the level of the lesion both in mouse and in man.
Collapse
Affiliation(s)
- Siti W. Mohd-Zin
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ahmed I. Marwan
- Laboratory for Fetal and Regenerative Biology, Colorado Fetal Care Center, Division of Pediatric Surgery, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, 12700 E 17th Ave, Aurora, CO 80045, USA
| | | | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noraishah M. Abdul-Aziz
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
57
|
Danno S, Kubouchi K, Mehruba M, Abe M, Natsume R, Sakimura K, Eguchi S, Oka M, Hirashima M, Yasuda H, Mukai H. PKN2 is essential for mouse embryonic development and proliferation of mouse fibroblasts. Genes Cells 2017; 22:220-236. [PMID: 28102564 DOI: 10.1111/gtc.12470] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/21/2016] [Indexed: 12/17/2022]
Abstract
PKN2, a member of the protein kinase N (PKN) family, has been suggested by in vitro culture cell experiments to bind to Rho/Rac GTPases and contributes to cell-cell contact and cell migration. To unravel the in vivo physiological function of PKN2, we targeted the PKN2 gene. Constitutive disruption of the mouse PKN2 gene resulted in growth retardation and lethality before embryonic day (E) 10.5. PKN2-/- embryo did not undergo axial turning and showed insufficient closure of the neural tube. Mouse embryonic fibroblasts (MEFs) derived from PKN2-/- embryos at E9.5 failed to grow. Cre-mediated ablation of PKN2 in PKN2flox/flox MEFs obtained from E14.5 embryos showed impaired cell proliferation, and cell cycle analysis of these MEFs showed a decrease in S-phase population. Our results show that PKN2 is essential for mouse embryonic development and cell-autonomous proliferation of primary MEFs in culture. Comparison of the PKN2-/- phenotype with the phenotypes of PKN1 and PKN3 knockout strains suggests that PKN2 has distinct nonredundant functions in vivo, despite the structural similarity and evolutionary relationship among the three isoforms.
Collapse
Affiliation(s)
- Sally Danno
- Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Koji Kubouchi
- Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Mona Mehruba
- Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Rie Natsume
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Satoshi Eguchi
- Department of Medical Biology, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Masahiro Oka
- Division of Dermatology, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino-ku, Sendai, 983-8512, Japan
| | | | - Hiroki Yasuda
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Hideyuki Mukai
- Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| |
Collapse
|
58
|
Mousa SA, Muralidharan-Chari V, Davis PJ. Interface between Thrombosis, Inflammation, and Angiogenesis in Cancer Progression. ANTI-ANGIOGENESIS STRATEGIES IN CANCER THERAPEUTICS 2017:51-68. [DOI: 10.1016/b978-0-12-802576-5.00004-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
59
|
Tanabe LM, List K. The role of type II transmembrane serine protease-mediated signaling in cancer. FEBS J 2016; 284:1421-1436. [PMID: 27870503 DOI: 10.1111/febs.13971] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/29/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
Abstract
Pericellular proteases have long been implicated in carcinogenesis. Previous research focused on these proteins, primarily as extracellular matrix (ECM) protein-degrading enzymes which allowed cancer cells to breach the basement membrane and invade surrounding tissue. However, recently, there has been a shift in the view of cell surface proteases, including serine proteases, as proteolytic modifiers of particular targets, including growth factors and protease-activated receptors, which are critical for the activation of oncogenic signaling pathways. Of the 176 human serine proteases currently identified, a subset of 17, known as type II transmembrane serine proteases (TTSPs). Many have been shown to be relevant to cancer progression since they were first identified as a family around the turn of the century. To this end, altered expression of TTSPs appeared as a trademark of several tumor types. However, the substrates and underlying signaling pathways remained unclear. Localization of these proteins to the cell surface places them in the unique position to mediate signal transduction between the cell and its surrounding environment. Many of the TTSPs have already been shown to play key roles in processes such as postnatal development, tissue homeostasis, and tumor progression, which share overlapping molecular mechanisms. In this review, we summarize the current knowledge regarding the role of the TTSP family in pro-oncogenic signaling.
Collapse
Affiliation(s)
- Lauren M Tanabe
- Department of Pharmacology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Karin List
- Department of Pharmacology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
60
|
Natural Endogenous Human Matriptase and Prostasin Undergo Zymogen Activation via Independent Mechanisms in an Uncoupled Manner. PLoS One 2016; 11:e0167894. [PMID: 27936035 PMCID: PMC5148038 DOI: 10.1371/journal.pone.0167894] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
The membrane-associated serine proteases matriptase and prostasin are believed to function in close partnership. Their zymogen activation has been reported to be tightly coupled, either as a matriptase-initiated proteolytic cascade or through a mutually dependent mechanism involving the formation of a reciprocal zymogen activation complex. Here we show that this putative relationship may not apply in the context of human matriptase and prostasin. First, the tightly coupled proteolytic cascade between matriptase and prostasin might not occur when modest matriptase activation is induced by sphingosine 1-phospahte in human mammary epithelial cells. Second, prostasin is not required and/or involved in matriptase autoactivation because matriptase can undergo zymogen activation in cells that do not endogenously express prostasin. Third, matriptase is not required for and/or involved in prostasin activation, since activated prostasin can be detected in cells expressing no endogenous matriptase. Finally, matriptase and prostasin both undergo zymogen activation through an apparently un-coupled mechanism in cells endogenously expressing both proteases, such as in Caco-2 cells. In these human enterocytes, matriptase is detected primarily in the zymogen form and prostasin predominantly as the activated form, either in complexes with protease inhibitors or as the free active form. The negligible levels of prostasin zymogen with high levels of matriptase zymogen suggests that the reciprocal zymogen activation complex is likely not the mechanism for matriptase zymogen activation. Furthermore, high level prostasin activation still occurs in Caco-2 variants with reduced or absent matriptase expression, indicating that matriptase is not required and/or involved in prostasin zymogen activation. Collectively, these data suggest that any functional relationship between natural endogenous human matriptase and prostasin does not occur at the level of zymogen activation.
Collapse
|
61
|
Kanemaru A, Yamamoto K, Kawaguchi M, Fukushima T, Lin CY, Johnson MD, Camerer E, Kataoka H. Deregulated matriptase activity in oral squamous cell carcinoma promotes the infiltration of cancer-associated fibroblasts by paracrine activation of protease-activated receptor 2. Int J Cancer 2016; 140:130-141. [PMID: 27615543 DOI: 10.1002/ijc.30426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/24/2016] [Accepted: 09/05/2016] [Indexed: 12/29/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are known to contribute to cancer progression. We have reported that cell surface expression of hepatocyte growth factor activator inhibitor 1 (HAI-1) is decreased in invasive oral squamous cell carcinoma (OSCC) cells. This study examined if HAI-1-insufficiency contributes to CAF recruitment in OSCC. Serum-free conditioned medium (SFCM) from a human OSCC line (SAS) stimulated the migration of 3 human fibroblast cell lines, NB1RGB, MRC5 and KD. SFCM from HAI-1-knockdown SAS showed an additive effect on the migration of NB1RGB and MRC5, but not KD. SAS SFCM induced protease-activated receptor-2 (PAR-2) expression in NB1RGB and MRC5, but not in KD, and a PAR-2 antagonist blocked the stimulatory effect of HAI-1 knockdown on migration of the PAR-2 expressing cell lines. Moreover, HAI-1-deficient SFCM showed additive stimulatory effects on the migration of wild-type but not PAR-2-deficient mouse fibroblasts. Therefore, the enhanced migration induced by HAI-1-insufficiency was mediated by PAR-2 activation in fibroblasts. This activation resulted from the deregulation of the activity of matriptase, a PAR-2 agonist protease. HAI-1 may thus prevent CAF recruitment to OSCC by controlling matriptase activity. When HAI-1 expression is reduced on OSCC, matriptase may contribute to CAF accumulation by paracrine activation of fibroblast PAR-2. Immunohistochemical analysis of resected OSCC revealed increased PAR2-positive CAFs in 35% (33/95) of the cases studied. The increased PAR-2 positive CAFs tended to correlate with infiltrative histology of the invasion front and shorter disease-free survival of the patients.
Collapse
Affiliation(s)
- Ai Kanemaru
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Koji Yamamoto
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Tsuyoshi Fukushima
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Chen-Yong Lin
- School of Medicine, Lambardi Comprehensive Cancer Centre, Georgetown University, Washington, DC
| | - Michael D Johnson
- School of Medicine, Lambardi Comprehensive Cancer Centre, Georgetown University, Washington, DC
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| |
Collapse
|
62
|
Iimura A, Yamazaki F, Suzuki T, Endo T, Nishida E, Kusakabe M. The E3 ubiquitin ligase Hace1 is required for early embryonic development in Xenopus laevis. BMC DEVELOPMENTAL BIOLOGY 2016; 16:31. [PMID: 27653971 PMCID: PMC5031333 DOI: 10.1186/s12861-016-0132-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 08/25/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (HACE1) regulates a wide variety of cellular processes. It has been shown that one of the targets of HACE1 is the GTP-bound form of the small GTPase Rac1. However, the role of HACE1 in early development remains unknown. RESULTS In situ hybridization revealed that Xenopus laevis hace1 is specifically expressed in the ectoderm at the blastula and gastrula stages and in the epidermis, branchial arch, kidney, and central nervous system at the tailbud stage. Knockdown of hace1 in Xenopus laevis embryos via antisense morpholino oligonucleotides led to defects in body axis elongation, pigment formation, and eye formation at the tadpole stage. Experiments with Keller sandwich explants showed that hace1 knockdown inhibited convergent extension, a morphogenetic movement known to be crucial for body axis elongation. In addition, time lapse imaging of whole embryos during the neurula stage indicated that hace1 knockdown also delayed neural tube closure. The defects caused by hace1 knockdown were partly rescued by knockdown of rac1. Moreover, embryos expressing a constitutively active form of Rac1 displayed phenotypes similar to those of embryos with hace1 knocked down. CONCLUSIONS Our results suggest that Xenopus laevis hace1 plays an important role in early embryonic development, possibly via regulation of Rac1 activity.
Collapse
Affiliation(s)
- Akira Iimura
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502 Japan
| | - Fuhito Yamazaki
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502 Japan
| | - Toshiyasu Suzuki
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502 Japan
| | - Tatsuya Endo
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502 Japan
| | - Eisuke Nishida
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502 Japan
| | - Morioh Kusakabe
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502 Japan
| |
Collapse
|
63
|
Nasri I, Bonnet D, Zwarycz B, d'Aldebert E, Khou S, Mezghani-Jarraya R, Quaranta M, Rolland C, Bonnart C, Mas E, Ferrand A, Cenac N, Magness S, Van Landeghem L, Vergnolle N, Racaud-Sultan C. PAR2-dependent activation of GSK3β regulates the survival of colon stem/progenitor cells. Am J Physiol Gastrointest Liver Physiol 2016; 311:G221-36. [PMID: 27313176 PMCID: PMC5007290 DOI: 10.1152/ajpgi.00328.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 05/16/2016] [Indexed: 01/31/2023]
Abstract
Protease-activated receptors PAR1 and PAR2 play an important role in the control of epithelial cell proliferation and migration. However, the survival of normal and tumor intestinal stem/progenitor cells promoted by proinflammatory mediators may be critical in oncogenesis. The glycogen synthase kinase-3β (GSK3β) pathway is overactivated in colon cancer cells and promotes their survival and drug resistance. We thus aimed to determine PAR1 and PAR2 effects on normal and tumor intestinal stem/progenitor cells and whether they involved GSK3β. First, PAR1 and PAR2 were identified in colon stem/progenitor cells by immunofluorescence. In three-dimensional cultures of murine crypt units or single tumor Caco-2 cells, PAR2 activation decreased numbers and size of normal or cancerous spheroids, and PAR2-deficient spheroids showed increased proliferation, indicating that PAR2 represses proliferation. PAR2-stimulated normal cells were more resistant to stress (serum starvation or spheroid passaging), suggesting prosurvival effects of PAR2 Accordingly, active caspase-3 was strongly increased in PAR2-deficient normal spheroids. PAR2 but not PAR1 triggered GSK3β activation through serine-9 dephosphorylation in normal and tumor cells. The PAR2-triggered GSK3β activation implicates an arrestin/PP2A/GSK3β complex that is dependent on the Rho kinase activity. Loss of PAR2 was associated with high levels of GSK3β nonactive form, strengthening the role of PAR2 in GSK3β activation. GSK3 pharmacological inhibition impaired the survival of PAR2-stimulated spheroids and serum-starved cells. Altogether our data identify PAR2/GSK3β as a novel pathway that plays a critical role in the regulation of stem/progenitor cell survival and proliferation in normal colon crypts and colon cancer.
Collapse
Affiliation(s)
- Imen Nasri
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,2Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia;
| | - Delphine Bonnet
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,3Service de Médecine Interne, Fédération Digestive, Centre Hospitalier Universitaire Purpan, Toulouse, France;
| | - Bailey Zwarycz
- 4Departments of Medicine and Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - Emilie d'Aldebert
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Sokchea Khou
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Raoudha Mezghani-Jarraya
- 2Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia;
| | - Muriel Quaranta
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Corinne Rolland
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Chrystelle Bonnart
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Emmanuel Mas
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,5Service de Gastroentérologie, Hépatologie et Nutrition, Hôpital des Enfants, Toulouse, France; and
| | - Audrey Ferrand
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Nicolas Cenac
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Scott Magness
- 4Departments of Medicine and Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - Laurianne Van Landeghem
- 6Institut National de la Santé et de la Recherche Médicale U913, Université de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Nathalie Vergnolle
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Claire Racaud-Sultan
- Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| |
Collapse
|
64
|
Szabo R, Lantsman T, Peters DE, Bugge TH. Delineation of proteolytic and non-proteolytic functions of the membrane-anchored serine protease prostasin. Development 2016; 143:2818-28. [PMID: 27385010 DOI: 10.1242/dev.137968] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/14/2016] [Indexed: 11/20/2022]
Abstract
The membrane-anchored serine proteases prostasin (PRSS8) and matriptase (ST14) initiate a cell surface proteolytic pathway essential for epithelial function. Mice expressing only catalytically inactive prostasin are viable, unlike prostasin null mice, indicating that at least some prostasin functions are non-proteolytic. Here we used knock-in mice expressing catalytically inactive prostasin (Prss8(Ki/Ki)) to show that the physiological and pathological functions of prostasin vary in their dependence on its catalytic activity. Whereas prostasin null mice exhibited partial embryonic and complete perinatal lethality, Prss8(Ki/Ki) mice displayed normal prenatal and postnatal survival. Unexpectedly, catalytically inactive prostasin caused embryonic lethality in mice lacking its cognate inhibitors HAI-1 (SPINT1) or HAI-2 (SPINT2). Proteolytically inactive prostasin, unlike the wild-type protease, was unable to activate matriptase during placentation. Surprisingly, all essential functions of prostasin in embryonic and postnatal development were compensated for by loss of HAI-1, indicating that prostasin is only required for mouse development and overall viability in the presence of this inhibitor. This study expands our knowledge of non-proteolytic functions of membrane-anchored serine proteases and provides unexpected new data on the mechanistic interactions between matriptase and prostasin in the context of epithelial development.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Taliya Lantsman
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diane E Peters
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA Program of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA 02110, USA
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
65
|
Ray HJ, Niswander LA. Dynamic behaviors of the non-neural ectoderm during mammalian cranial neural tube closure. Dev Biol 2016; 416:279-85. [PMID: 27343896 DOI: 10.1016/j.ydbio.2016.06.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/26/2016] [Accepted: 06/19/2016] [Indexed: 11/27/2022]
Abstract
The embryonic brain and spinal cord initially form through the process of neural tube closure (NTC). NTC is thought to be highly similar between rodents and humans, and studies of mouse genetic mutants have greatly increased our understanding of the molecular basis of NTC with relevance for human neural tube defects. In addition, studies using amphibian and chick embryos have shed light into the cellular and tissue dynamics underlying NTC. However, the dynamics of mammalian NTC has been difficult to study due to in utero development until recently when advances in mouse embryo ex vivo culture techniques along with confocal microscopy have allowed for imaging of mouse NTC in real time. Here, we have performed live imaging of mouse embryos with a particular focus on the non-neural ectoderm (NNE). Previous studies in multiple model systems have found that the NNE is important for proper NTC, but little is known about the behavior of these cells during mammalian NTC. Here we utilized a NNE-specific genetic labeling system to assess NNE dynamics during murine NTC and identified different NNE cell behaviors as the cranial region undergoes NTC. These results bring valuable new insight into regional differences in cellular behavior during NTC that may be driven by different molecular regulators and which may underlie the various positional disruptions of NTC observed in humans with neural tube defects.
Collapse
Affiliation(s)
- Heather J Ray
- Department of Pediatrics, Cell Biology Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Lee A Niswander
- Department of Pediatrics, Cell Biology Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
66
|
Rolo A, Savery D, Escuin S, de Castro SC, Armer HEJ, Munro PMG, Molè MA, Greene NDE, Copp AJ. Regulation of cell protrusions by small GTPases during fusion of the neural folds. eLife 2016; 5:e13273. [PMID: 27114066 PMCID: PMC4846376 DOI: 10.7554/elife.13273] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/18/2016] [Indexed: 11/26/2022] Open
Abstract
Epithelial fusion is a crucial process in embryonic development, and its failure underlies several clinically important birth defects. For example, failure of neural fold fusion during neurulation leads to open neural tube defects including spina bifida. Using mouse embryos, we show that cell protrusions emanating from the apposed neural fold tips, at the interface between the neuroepithelium and the surface ectoderm, are required for completion of neural tube closure. By genetically ablating the cytoskeletal regulators Rac1 or Cdc42 in the dorsal neuroepithelium, or in the surface ectoderm, we show that these protrusions originate from surface ectodermal cells and that Rac1 is necessary for the formation of membrane ruffles which typify late closure stages, whereas Cdc42 is required for the predominance of filopodia in early neurulation. This study provides evidence for the essential role and molecular regulation of membrane protrusions prior to fusion of a key organ primordium in mammalian development. DOI:http://dx.doi.org/10.7554/eLife.13273.001 The neural tube is an embryonic structure that gives rise to the brain and spinal cord. It originates from a flat sheet of cells – the neural plate – that rolls up and fuses to form a tube during development. If this closure fails, it leads to birth defects such as spina bifida, a condition that causes severe disability because babies are born with an exposed and damaged spinal cord. As the edges of the neural plate meet, they need to fuse together to produce a closed tube. It was known that cells at these edges extend protrusions. However, it was unclear how these protrusions are regulated, whether they arise from neural or non-neural cells and whether or not they are required for the neural tube to close fully. By studying mutant mouse embryos, Rolo et al. found that cellular protrusions are indeed required for the neural tube to close completely. These protrusions proved to be regulated by proteins called Rac1 and Cdc42, which control the filaments inside the cell that are responsible for cell shape and movement. Rolo et al. also found that the cells that give rise to the protrusions are not part of the neural plate itself. Instead, these cells are neighboring cells from the layer that later forms the epidermis of the skin (the surface ectoderm). Future studies will need to investigate which signals instruct those precise cells to make protrusions and to discover what happens to the protrusions after contact is made with cells on the opposite side. It will also be important to determine whether spina bifida may arise in humans if the protrusions are defective or absent. DOI:http://dx.doi.org/10.7554/eLife.13273.002
Collapse
Affiliation(s)
- Ana Rolo
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Dawn Savery
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Sarah Escuin
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Sandra C de Castro
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Hannah E J Armer
- Imaging Unit, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Peter M G Munro
- Imaging Unit, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Matteo A Molè
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
67
|
Matriptase activation connects tissue factor-dependent coagulation initiation to epithelial proteolysis and signaling. Blood 2016; 127:3260-9. [PMID: 27114461 DOI: 10.1182/blood-2015-11-683110] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/11/2016] [Indexed: 12/23/2022] Open
Abstract
The coagulation cascade is designed to sense tissue injury by physical separation of the membrane-anchored cofactor tissue factor (TF) from inactive precursors of coagulation proteases circulating in plasma. Once TF on epithelial and other extravascular cells is exposed to plasma, sequential activation of coagulation proteases coordinates hemostasis and contributes to host defense and tissue repair. Membrane-anchored serine proteases (MASPs) play critical roles in the development and homeostasis of epithelial barrier tissues; how MASPs are activated in mature epithelia is unknown. We here report that proteases of the extrinsic pathway of blood coagulation transactivate the MASP matriptase, thus connecting coagulation initiation to epithelial proteolysis and signaling. Exposure of TF-expressing cells to factors (F) VIIa and Xa triggered the conversion of latent pro-matriptase to an active protease, which in turn cleaved the pericellular substrates protease-activated receptor-2 (PAR2) and pro-urokinase. An activation pathway-selective PAR2 mutant resistant to direct cleavage by TF:FVIIa and FXa was activated by these proteases when cells co-expressed pro-matriptase, and matriptase transactivation was necessary for efficient cleavage and activation of wild-type PAR2 by physiological concentrations of TF:FVIIa and FXa. The coagulation initiation complex induced rapid and prolonged enhancement of the barrier function of epithelial monolayers that was dependent on matriptase transactivation and PAR2 signaling. These observations suggest that the coagulation cascade engages matriptase to help coordinate epithelial defense and repair programs after injury or infection, and that matriptase may contribute to TF-driven pathogenesis in cancer and inflammation.
Collapse
|
68
|
Morla L, Edwards A, Crambert G. New insights into sodium transport regulation in the distal nephron: Role of G-protein coupled receptors. World J Biol Chem 2016; 7:44-63. [PMID: 26981195 PMCID: PMC4768124 DOI: 10.4331/wjbc.v7.i1.44] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/02/2015] [Accepted: 11/25/2015] [Indexed: 02/05/2023] Open
Abstract
The renal handling of Na+ balance is a major determinant of the blood pressure (BP) level. The inability of the kidney to excrete the daily load of Na+ represents the primary cause of chronic hypertension. Among the different segments that constitute the nephron, those present in the distal part (i.e., the cortical thick ascending limb, the distal convoluted tubule, the connecting and collecting tubules) play a central role in the fine-tuning of renal Na+ excretion and are the target of many different regulatory processes that modulate Na+ retention more or less efficiently. G-protein coupled receptors (GPCRs) are crucially involved in this regulation and could represent efficient pharmacological targets to control BP levels. In this review, we describe both classical and novel GPCR-dependent regulatory systems that have been shown to modulate renal Na+ absorption in the distal nephron. In addition to the multiplicity of the GPCR that regulate Na+ excretion, this review also highlights the complexity of these different pathways, and the connections between them.
Collapse
|
69
|
Ray HJ, Niswander LA. Grainyhead-like 2 downstream targets act to suppress epithelial-to-mesenchymal transition during neural tube closure. Development 2016; 143:1192-204. [PMID: 26903501 DOI: 10.1242/dev.129825] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/16/2016] [Indexed: 12/29/2022]
Abstract
The transcription factor grainyhead-like 2 (GRHL2) is expressed in non-neural ectoderm (NNE) and Grhl2 loss results in fully penetrant cranial neural tube defects (NTDs) in mice. GRHL2 activates expression of several epithelial genes; however, additional molecular targets and functional processes regulated by GRHL2 in the NNE remain to be determined, as well as the underlying cause of the NTDs in Grhl2 mutants. Here, we find that Grhl2 loss results in abnormal mesenchymal phenotypes in the NNE, including aberrant vimentin expression and increased cellular dynamics that affects the NNE and neural crest cells. The resulting loss of NNE integrity contributes to an inability of the cranial neural folds to move toward the midline and results in NTD. Further, we identified Esrp1, Sostdc1, Fermt1, Tmprss2 and Lamc2 as novel NNE-expressed genes that are downregulated in Grhl2 mutants. Our in vitro assays show that they act as suppressors of the epithelial-to-mesenchymal transition (EMT). Thus, GRHL2 promotes the epithelial nature of the NNE during the dynamic events of neural tube formation by both activating key epithelial genes and actively suppressing EMT through novel downstream EMT suppressors.
Collapse
Affiliation(s)
- Heather J Ray
- Department of Pediatrics, Cell Biology Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Lee A Niswander
- Department of Pediatrics, Cell Biology Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| |
Collapse
|
70
|
Walentin K, Hinze C, Schmidt-Ott KM. The basal chorionic trophoblast cell layer: An emerging coordinator of placenta development. Bioessays 2016; 38:254-65. [PMID: 26778584 DOI: 10.1002/bies.201500087] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
During gestation, fetomaternal exchange occurs in the villous tree (labyrinth) of the placenta. Development of this structure depends on tightly coordinated cellular processes of branching morphogenesis and differentiation of specialized trophoblast cells. The basal chorionic trophoblast (BCT) cell layer that localizes next to the chorioallantoic interface is of critical importance for labyrinth morphogenesis in rodents. Gcm1-positive cell clusters within this layer initiate branching morphogenesis thereby guiding allantoic fetal blood vessels towards maternal blood sinuses. Later these cells differentiate and contribute to the syncytiotrophoblast of the fetomaternal barrier. Additional cells within the BCT layer sustain continued morphogenesis, possibly through a repopulating progenitor population. Several mouse mutants highlight the importance of a structurally intact BCT epithelium, and a growing number of studies addresses its patterning and epithelial architecture. Here, we review and discuss emerging concepts in labyrinth development focussing on the biology of the BCT cell layer.
Collapse
Affiliation(s)
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
71
|
Abstract
Membrane-anchored serine proteases are a group of extracellular serine proteases tethered directly to plasma membranes, via a C-terminal glycosylphosphatidylinositol linkage (GPI-anchored), a C-terminal transmembrane domain (Type I), or an N-terminal transmembrane domain (Type II). A variety of biochemical, cellular, and in vivo studies have established that these proteases are important pericellular contributors to processes vital for the maintenance of homeostasis, including food digestion, blood pressure regulation, hearing, epithelial permeability, sperm maturation, and iron homeostasis. These enzymes are hijacked by viruses to facilitate infection and propagation, and their misregulation is associated with a wide range of diseases, including cancer malignancy.
Collapse
|
72
|
Chai AC, Robinson AL, Chai KX, Chen LM. Ibuprofen regulates the expression and function of membrane-associated serine proteases prostasin and matriptase. BMC Cancer 2015; 15:1025. [PMID: 26715240 PMCID: PMC4696080 DOI: 10.1186/s12885-015-2039-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/21/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The glycosylphosphatidylinositol-anchored extracellular membrane serine protease prostasin is expressed in normal bladder urothelial cells. Bladder inflammation reduces prostasin expression and a loss of prostasin expression is associated with epithelial-mesenchymal transition (EMT) in human bladder transitional cell carcinomas. Non-steroidal anti-inflammatory drugs (NSAIDs) decrease the incidence of various cancers including bladder cancer, but the molecular mechanisms underlying the anticancer effect of NSAIDs are not fully understood. METHODS The normal human bladder urothelial cell line UROtsa, the normal human trophoblast cell line B6Tert-1, human bladder transitional cell carcinoma cell lines UM-UC-5 and UM-UC-9, and the human breast cancer cell line JIMT-1 were used for the study. Expression changes of the serine proteases prostasin and matriptase, and cyclooxygenases (COX-1 and COX-2) in these cells following ibuprofen treatments were analyzed by means of reverse-transcription/quantitative polymerase chain reaction (RT-qPCR) and immunoblotting. The functional role of the ibuprofen-regulated prostasin in epithelial tight junction formation and maintenance was assessed by measuring the transepithelial electrical resistance (TEER) and epithelial permeability in the B6Tert-1 cells. Prostasin's effects on tight junctions were also evaluated in B6Tert-1 cells over-expressing a recombinant human prostasin, silenced for prostasin expression, or treated with a functionally-blocking prostasin antibody. Matriptase zymogen activation was examined in cells over-expressing prostasin. RESULTS Ibuprofen increased prostasin expression in the UROtsa and the B6Tert-1 cells. Cyclooxygenase-2 (COX-2) expression was up-regulated at both the mRNA and the protein levels in the UROtsa cells by ibuprofen in a dose-dependent manner, but was not a requisite for up-regulating prostasin expression. The ibuprofen-induced prostasin contributed to the formation and maintenance of the epithelial tight junctions in the B6Tert-1 cells. The matriptase zymogen was down-regulated in the UROtsa cells by ibuprofen possibly as a result of the increased prostasin expression because over-expressing prostasin leads to matriptase activation and zymogen down-regulation in the UROtsa, JIMT-1, and B6Tert-1 cells. The expression of prostasin and matriptase was differentially regulated by ibuprofen in the bladder cancer cells. CONCLUSIONS Ibuprofen has been suggested for use in treating bladder cancer. Our results bring the epithelial extracellular membrane serine proteases prostasin and matriptase into the potential molecular mechanisms of the anticancer effect of NSAIDs.
Collapse
Affiliation(s)
- Andreas C Chai
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4000 Central Florida Boulevard, Building 20, Room 323, Orlando, FL, 32816-2364, USA
| | - Andrew L Robinson
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4000 Central Florida Boulevard, Building 20, Room 323, Orlando, FL, 32816-2364, USA
| | - Karl X Chai
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4000 Central Florida Boulevard, Building 20, Room 323, Orlando, FL, 32816-2364, USA
| | - Li-Mei Chen
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4000 Central Florida Boulevard, Building 20, Room 323, Orlando, FL, 32816-2364, USA.
| |
Collapse
|
73
|
La matriptase, un activateur de PAR2 dans le néphron distal ? Nephrol Ther 2015. [DOI: 10.1016/j.nephro.2015.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
74
|
Abstract
In the last decade, several mouse models for RhoA, Rac1, and Cdc42 have emerged and have contributed a great deal to understanding the precise functions of Rho GTPases at early stages of development. This review summarizes our current knowledge of various mouse models of tissue-specific ablation of Cdc42, Rac1, and RhoA with emphasis on early embryogenesis, epithelial and skin morphogenesis, tubulogenesis, development of the central nervous system, and limb development.
Collapse
Affiliation(s)
- Philippe M Duquette
- a McGill University ; Department of Anatomy and Cell Biology ; Montreal , QC Canada
| | | |
Collapse
|
75
|
A balance between TFPI and thrombin-mediated platelet activation is required for murine embryonic development. Blood 2015; 125:4078-84. [PMID: 25954015 DOI: 10.1182/blood-2015-03-633958] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/03/2015] [Indexed: 11/20/2022] Open
Abstract
Tissue factor pathway inhibitor (TFPI) is a critical anticoagulant protein present in endothelium and platelets. Mice lacking TFPI (Tfpi(-/-)) die in utero from disseminated intravascular coagulation. They are rescued by concomitant tissue factor (TF) deficiency, demonstrating that TFPI modulates TF function in vivo. Recent studies have found TFPI inhibits prothrombinase activity during the initiation of coagulation and limits platelet accumulation during thrombus formation, implicating TFPI in modulating platelet procoagulant activity. To examine whether altered platelet function would compensate for the lack of TFPI and rescue TFPI-null embryonic lethality, Tfpi(+/-) mice lacking the platelet thrombin receptor, protease activated receptor 4 (PAR4; Par4(-/-)), or its coreceptor, PAR3, were mated. PAR3 deficiency did not rescue Tfpi(-/-) embryos, but >40% of expected Tfpi(-/-):Par4(-/-) offspring survived to adulthood. Adult Tfpi(-/-):Par4(-/-) mice did not exhibit overt thrombosis. However, they had focal sterile inflammation with fibrin(ogen) deposition in the liver and elevated plasma thrombin-antithrombin complexes, indicating activation of coagulation at baseline. Tfpi(-/-):Par4(-/-) mice have platelet and fibrin accumulation similar to Par4(-/-) mice following venous electrolytic injury but were more susceptible than Par4(-/-) mice to TF-induced pulmonary embolism. In addition, ∼30% of the Tfpi(-/-):Par4(-/-) mice were born with short tails. Tfpi(-/-):Par4(-/-) mice are the first adult mice described that lack TFPI with unaltered TF. They demonstrate that TFPI physiologically modulates thrombin-dependent platelet activation in a manner that is required for successful embryonic development and identify a role for TFPI in dampening intravascular procoagulant stimuli that lead to thrombin generation, even in the absence of thrombin-mediated platelet activation.
Collapse
|
76
|
Hashimoto H, Robin FB, Sherrard KM, Munro EM. Sequential contraction and exchange of apical junctions drives zippering and neural tube closure in a simple chordate. Dev Cell 2015; 32:241-55. [PMID: 25625209 DOI: 10.1016/j.devcel.2014.12.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 11/07/2014] [Accepted: 12/21/2014] [Indexed: 10/24/2022]
Abstract
Unidirectional zippering is a key step in neural tube closure that remains poorly understood. Here, we combine experimental and computational approaches to identify the mechanism for zippering in a basal chordate, Ciona intestinalis. We show that myosin II is activated sequentially from posterior to anterior along the neural/epidermal (Ne/Epi) boundary just ahead of the advancing zipper. This promotes rapid shortening of Ne/Epi junctions, driving the zipper forward and drawing the neural folds together. Cell contact rearrangements (Ne/Epi + Ne/Epi → Ne/Ne + Epi/Epi) just behind the zipper lower tissue resistance to zipper progression by allowing transiently stretched cells to detach and relax toward isodiametric shapes. Computer simulations show that measured differences in junction tension, timing of primary contractions, and delay before cell detachment are sufficient to explain the speed and direction of zipper progression and highlight key advantages of a sequential contraction mechanism for robust efficient zippering.
Collapse
Affiliation(s)
- Hidehiko Hashimoto
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Francois B Robin
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA.
| | - Kristin M Sherrard
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Edwin M Munro
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA; Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
77
|
Kimura-Yoshida C, Mochida K, Ellwanger K, Niehrs C, Matsuo I. Fate Specification of Neural Plate Border by Canonical Wnt Signaling and Grhl3 is Crucial for Neural Tube Closure. EBioMedicine 2015; 2:513-27. [PMID: 26288816 PMCID: PMC4535158 DOI: 10.1016/j.ebiom.2015.04.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 12/05/2022] Open
Abstract
During primary neurulation, the separation of a single-layered ectodermal sheet into the surface ectoderm (SE) and neural tube specifies SE and neural ectoderm (NE) cell fates. The mechanisms underlying fate specification in conjunction with neural tube closure are poorly understood. Here, by comparing expression profiles between SE and NE lineages, we observed that uncommitted progenitor cells, expressing stem cell markers, are present in the neural plate border/neural fold prior to neural tube closure. Our results also demonstrated that canonical Wnt and its antagonists, DKK1/KREMEN1, progressively specify these progenitors into SE or NE fates in accord with the progress of neural tube closure. Additionally, SE specification of the neural plate border via canonical Wnt signaling is directed by the grainyhead-like 3 (Grhl3) transcription factor. Thus, we propose that the fate specification of uncommitted progenitors in the neural plate border by canonical Wnt signaling and its downstream effector Grhl3 is crucial for neural tube closure. This study implicates that failure in critical genetic factors controlling fate specification of progenitor cells in the neural plate border/neural fold coordinated with neural tube closure may be potential causes of human neural tube defects. Neural plate border/neural fold possesses stem cell-like characters during primary neurulation. Canonical Wnt and its antagonists progressively specify progenitors into surface or neural fates upon neural tube closure. Fate specification into surface ectoderm in the neural fold is directed by the Grhl3 transcription factor. Fate specification of uncommitted progenitors in the neural plate border is intimately coupled to neural tube closure.
Collapse
Affiliation(s)
- Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kyoko Mochida
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kristina Ellwanger
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany ; Institute of Molecular Biology, 55128 Mainz, Germany
| | - Isao Matsuo
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
78
|
Hepatocyte growth factor activator inhibitor type 1 maintains the assembly of keratin into desmosomes in keratinocytes by regulating protease-activated receptor 2-dependent p38 signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1610-23. [PMID: 25842366 DOI: 10.1016/j.ajpath.2015.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/26/2015] [Accepted: 02/03/2015] [Indexed: 11/23/2022]
Abstract
Hepatocyte growth factor activator inhibitor type 1 (HAI-1; official symbol SPINT1) is a membrane-associated serine proteinase inhibitor abundantly expressed in epithelial tissues. Genetically engineered mouse models demonstrated that HAI-1 is critical for epidermal function, possibly through direct and indirect regulation of cell surface proteases, such as matriptase and prostasin. To obtain a better understanding of the role of HAI-1 in maintaining epidermal integrity, we performed ultrastructural analysis of Spint1-deleted mouse epidermis and organotypic culture of an HAI-1 knockdown (KD) human keratinocyte cell line, HaCaT. We found that the aggregation of tonofilaments to desmosomes was significantly reduced in HAI-1-deficient mouse epidermis with decreased desmosome number. Similar findings were observed in HAI-1 KD HaCaT organotypic cultures. Immunoblot and immunohistochemical analyses revealed that p38 mitogen-activated protein kinase was activated in response to HAI-1 insufficiency. Treatment of HAI-1 KD HaCaT cells with a p38 inhibitor abrogated the above-observed ultrastructural abnormalities. The activation of p38 induced by the loss of HAI-1 likely resulted from enhanced signaling of protease-activated receptor-2 (PAR-2), because its silencing abrogated the enhanced activation of p38. Consequently, treatment of HAI-1 KD HaCaT cells with a serine protease inhibitor, aprotinin, or PAR-2 antagonist alleviated the abnormal ultrastructural phenotype in organotypic culture. These results suggest that HAI-1 may have a critical role in maintaining normal keratinocyte morphology through regulation of PAR-2-dependent p38 mitogen-activated protein kinase signaling.
Collapse
|
79
|
Abstract
Neural tube defects (NTDs), including spina bifida and anencephaly, are severe birth defects of the central nervous system that originate during embryonic development when the neural tube fails to close completely. Human NTDs are multifactorial, with contributions from both genetic and environmental factors. The genetic basis is not yet well understood, but several nongenetic risk factors have been identified as have possibilities for prevention by maternal folic acid supplementation. Mechanisms underlying neural tube closure and NTDs may be informed by experimental models, which have revealed numerous genes whose abnormal function causes NTDs and have provided details of critical cellular and morphological events whose regulation is essential for closure. Such models also provide an opportunity to investigate potential risk factors and to develop novel preventive therapies.
Collapse
Affiliation(s)
- Nicholas D E Greene
- Newlife Birth Defects Research Center, Institute of Child Health, University College London, WC1N 1EH, United Kingdom;
| | | |
Collapse
|
80
|
Tudpor K, van der Eerden BCJ, Jongwattanapisan P, Roelofs JJTH, van Leeuwen JPTM, Bindels RJM, Hoenderop JGJ. Thrombin receptor deficiency leads to a high bone mass phenotype by decreasing the RANKL/OPG ratio. Bone 2015; 72:14-22. [PMID: 25460576 DOI: 10.1016/j.bone.2014.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/26/2014] [Accepted: 11/10/2014] [Indexed: 12/25/2022]
Abstract
Thrombin and its receptor (TR) are, respectively, expressed in osteoclasts and osteoblasts. However, their physiological roles on bone metabolism have not been fully elucidated. Here we investigated the bone microarchitecture by micro-computed tomography (μCT) and demonstrated increased trabecular and cortical bone mass in femurs of TR KO mice compared to WT littermates. Trabecular thickness and connectivity were significantly enhanced. The physiological role of TR on both inorganic and organic phases of bone is illustrated by a significant increase in BMD and a decrease in urinary deoxypyridinoline (DPD) crosslink concentration in TR KO mice. Moreover, TR KO cortical bone expanded and had a higher polar moment of inertia (J), implying stronger bone. Bone histomorphometry illustrated unaltered osteoblast and osteoclast number and surface in femoral metaphyses, indicating that thrombin/TR regulates osteoblasts and osteoclasts at functional levels. Serum analysis showed a decrease in RANKL and an increase in osteoprotegerin (OPG) levels and reflected a reduced RANKL/OPG ratio in the TR KO group. In vitro experiments using MC3T3 pre-osteoblasts demonstrated a TR-dependent stimulatory effect of thrombin on the RANKL/OPG ratio. This effect was blocked by TR antagonist and p42/p44-ERK inhibitor. In addition, thrombin also intensified p42/p44-ERK expression and phosphorylation. In conclusion, the thrombin/TR system maintains normal bone remodeling by activating RANKL and limiting OPG synthesis by osteoblasts through the p42/44-ERK signaling pathway. Consequently, TR deficiency inhibits osteoclastogenesis, resulting in a high bone mass phenotype.
Collapse
Affiliation(s)
- Kukiat Tudpor
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands
| | | | - Prapaporn Jongwattanapisan
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands.
| |
Collapse
|
81
|
Mao X, Del Bigio MR. Interference with protease-activated receptor 1 does not reduce damage to subventricular zone cells of immature rodent brain following exposure to blood or blood plasma. J Negat Results Biomed 2015; 14:3. [PMID: 25649264 PMCID: PMC4327806 DOI: 10.1186/s12952-014-0022-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/22/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Prior work showed that whole blood, plasma, and serum injections are damaging to the neonatal rodent brain in a model of intracerebral/periventricular hemorrhage. Thrombin alone is also damaging. In adult animal models of hemorrhagic stroke, the protease-activated (thrombin) receptor PAR1 mediates some of the brain damage. We hypothesized that PAR1 interference will reduce the adverse effects of blood products on immature rodent brain and cells. RESULTS Cultured oligodendrocyte precursor cells from rats and mice were exposed to blood plasma with and without the PAR1 antagonists SCH-79797 or BMS-200261. In concentrations previously shown to have activity on brain cells, neither drug showed evidence of protection against the toxicity of blood plasma. Newborn mice (wild type, heterozygous, and PAR1 knockout) were subjected to intracerebral injection of autologous whole blood into the periventricular region of the frontal lobe. Cell proliferation, measured by Ki67 immunoreactivity in the subventricular zone, was suppressed at 1 and 2 days, and was not normalized in the knockout mice. Cell apoptosis, measured by activated caspase 3 immunoreactivity, was not apparent in the subventricular zone. Increased apoptosis in periventricular striatal cells was not normalized in the knockout mice. CONCLUSION Interference with the thrombin-PAR1 system does not reduce the adverse effects of blood on germinal cells of the immature rodent brain. PAR1 interference is unlikely to be a useful treatment for reducing the brain damage that accompanies periventricular (germinal matrix) hemorrhage, a common complication of premature birth.
Collapse
Affiliation(s)
- Xiaoyan Mao
- Department of Pathology, University of Manitoba, and Children's Hospital Research Institute of Manitoba, 401 Brodie Centre, 715 McDermot Avenue, Winnipeg, MB, R3E 3P5, Canada.
| | - Marc R Del Bigio
- Department of Pathology, University of Manitoba, and Children's Hospital Research Institute of Manitoba, 401 Brodie Centre, 715 McDermot Avenue, Winnipeg, MB, R3E 3P5, Canada.
| |
Collapse
|
82
|
Driesbaugh KH, Buzza MS, Martin EW, Conway GD, Kao JPY, Antalis TM. Proteolytic activation of the protease-activated receptor (PAR)-2 by the glycosylphosphatidylinositol-anchored serine protease testisin. J Biol Chem 2014; 290:3529-41. [PMID: 25519908 DOI: 10.1074/jbc.m114.628560] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protease-activated receptors (PARs) are a family of seven-transmembrane, G-protein-coupled receptors that are activated by multiple serine proteases through specific N-terminal proteolytic cleavage and the unmasking of a tethered ligand. The majority of PAR-activating proteases described to date are soluble proteases that are active during injury, coagulation, and inflammation. Less investigation, however, has focused on the potential for membrane-anchored serine proteases to regulate PAR activation. Testisin is a unique trypsin-like serine protease that is tethered to the extracellular membrane of cells through a glycophosphatidylinositol (GPI) anchor. Here, we show that the N-terminal domain of PAR-2 is a substrate for testisin and that proteolytic cleavage of PAR-2 by recombinant testisin activates downstream signaling pathways, including intracellular Ca(2+) mobilization and ERK1/2 phosphorylation. When testisin and PAR-2 are co-expressed in HeLa cells, GPI-anchored testisin specifically releases the PAR-2 tethered ligand. Conversely, knockdown of endogenous testisin in NCI/ADR-Res ovarian tumor cells reduces PAR-2 N-terminal proteolytic cleavage. The cleavage of PAR-2 by testisin induces activation of the intracellular serum-response element and NFκB signaling pathways and the induction of IL-8 and IL-6 cytokine gene expression. Furthermore, the activation of PAR-2 by testisin results in the loss and internalization of PAR-2 from the cell surface. This study reveals a new biological substrate for testisin and is the first demonstration of the activation of a PAR by a serine protease GPI-linked to the cell surface.
Collapse
Affiliation(s)
- Kathryn H Driesbaugh
- From the Department of Physiology, Center for Vascular and Inflammatory Diseases, and
| | - Marguerite S Buzza
- From the Department of Physiology, Center for Vascular and Inflammatory Diseases, and
| | - Erik W Martin
- From the Department of Physiology, Center for Vascular and Inflammatory Diseases, and
| | - Gregory D Conway
- From the Department of Physiology, Center for Vascular and Inflammatory Diseases, and
| | - Joseph P Y Kao
- From the Department of Physiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Toni M Antalis
- From the Department of Physiology, Center for Vascular and Inflammatory Diseases, and
| |
Collapse
|
83
|
Gordon WM, Zeller MD, Klein RH, Swindell WR, Ho H, Espetia F, Gudjonsson JE, Baldi PF, Andersen B. A GRHL3-regulated repair pathway suppresses immune-mediated epidermal hyperplasia. J Clin Invest 2014; 124:5205-18. [PMID: 25347468 DOI: 10.1172/jci77138] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/18/2014] [Indexed: 12/27/2022] Open
Abstract
Dermal infiltration of T cells is an important step in the onset and progression of immune-mediated skin diseases such as psoriasis; however, it is not known whether epidermal factors play a primary role in the development of these diseases. Here, we determined that the prodifferentiation transcription factor grainyhead-like 3 (GRHL3), which is essential during epidermal development, is dispensable for adult skin homeostasis, but required for barrier repair after adult epidermal injury. Consistent with activation of a GRHL3-regulated repair pathway in psoriasis, we found that GRHL3 is upregulated in lesional skin and binds known epidermal differentiation gene targets. Using an imiquimod-induced model of immune-mediated epidermal hyperplasia, we found that mice lacking GRHL3 have an exacerbated epidermal damage response, greater sensitivity to disease induction, delayed resolution of epidermal lesions, and resistance to anti-IL-22 therapy compared with WT animals. ChIP-Seq and gene expression profiling of murine skin revealed that while GRHL3 regulates differentiation pathways both during development and during repair from immune-mediated damage, it targets distinct sets of genes in the 2 processes. In particular, GRHL3 suppressed a number of alarmin and other proinflammatory genes after immune injury. This study identifies a GRHL3-regulated epidermal barrier repair pathway that suppresses disease initiation and helps resolve existing lesions in immune-mediated epidermal hyperplasia.
Collapse
|
84
|
TMPRSS13 deficiency impairs stratum corneum formation and epidermal barrier acquisition. Biochem J 2014; 461:487-95. [PMID: 24832573 DOI: 10.1042/bj20140337] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Membrane-anchored serine proteases serve as important regulators of multiple developmental and homoeostatic processes in mammals. TMPRSS13 (transmembrane protease, serine 13; also known as mosaic serine protease large-form, MSPL) is a membrane-anchored serine protease with unknown biological functions. In the present study, we used mice with the Tmprss13 gene disrupted by a β-galactosidase-neomycin fusion gene insertion to study the expression and function of the membrane-anchored serine protease. High levels of Tmprss13 expression were found in the epithelia of the oral cavity, upper digestive tract and skin. Compatible with this expression pattern, Tmprss13-deficient mice displayed abnormal skin development, leading to a compromised barrier function, as measured by the transepidermal fluid loss rate of newborn mice. The present study provides the first biological function for the transmembrane serine protease TMPRSS13.
Collapse
|
85
|
Crisante G, Battista L, Iwaszkiewicz J, Nesca V, Mérillat AM, Sergi C, Zoete V, Frateschi S, Hummler E. The CAP1/Prss8 catalytic triad is not involved in PAR2 activation and protease nexin-1 (PN-1) inhibition. FASEB J 2014; 28:4792-805. [PMID: 25138159 DOI: 10.1096/fj.14-253781] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Serine proteases, serine protease inhibitors, and protease-activated receptors (PARs) are responsible for several human skin disorders characterized by impaired epidermal permeability barrier function, desquamation, and inflammation. In this study, we addressed the consequences of a catalytically dead serine protease on epidermal homeostasis, the activation of PAR2 and the inhibition by the serine protease inhibitor nexin-1. The catalytically inactive serine protease CAP1/Prss8, when ectopically expressed in the mouse, retained the ability to induce skin disorders as well as its catalytically active counterpart (75%, n=81). Moreover, this phenotype was completely normalized in a PAR2-null background, indicating that the effects mediated by the catalytically inactive CAP1/Prss8 depend on PAR2 (95%, n=131). Finally, nexin-1 displayed analogous inhibitory capacity on both wild-type and inactive mutant CAP1/Prss8 in vitro and in vivo (64% n=151 vs. 89% n=109, respectively), indicating that the catalytic site of CAP1/Prss8 is dispensable for nexin-1 inhibition. Our results demonstrate a novel inhibitory interaction between CAP1/Prss8 and nexin-1, opening the search for specific CAP1/Prss8 antagonists that are independent of its catalytic activity.
Collapse
Affiliation(s)
| | | | - Justyna Iwaszkiewicz
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | | | | | - Chloé Sergi
- Department of Pharmacology and Toxicology and
| | - Vincent Zoete
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
86
|
Szabo R, Peters DE, Kosa P, Camerer E, Bugge TH. Regulation of feto-maternal barrier by matriptase- and PAR-2-mediated signaling is required for placental morphogenesis and mouse embryonic survival. PLoS Genet 2014; 10:e1004470. [PMID: 25078604 PMCID: PMC4117450 DOI: 10.1371/journal.pgen.1004470] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 05/14/2014] [Indexed: 11/19/2022] Open
Abstract
The development of eutherian mammalian embryos is critically dependent on the selective bi-directional transport of molecules across the placenta. Here, we uncover two independent and partially redundant protease signaling pathways that include the membrane-anchored serine proteases, matriptase and prostasin, and the G protein-coupled receptor PAR-2 that mediate the establishment of a functional feto-maternal barrier. Mice with a combined matriptase and PAR-2 deficiency do not survive to term and the survival of matriptase-deficient mice heterozygous for PAR-2 is severely diminished. Embryos with the combined loss of PAR-2 and matriptase or PAR-2 and the matriptase partner protease, prostasin, uniformly die on or before embryonic day 14.5. Despite the extensive co-localization of matriptase, prostasin, and PAR-2 in embryonic epithelia, the overall macroscopic and histological analysis of the double-deficient embryos did not reveal any obvious developmental abnormalities. In agreement with this, the conditional deletion of matriptase from the embryo proper did not affect the prenatal development or survival of PAR-2-deficient mice, indicating that the critical redundant functions of matriptase/prostasin and PAR-2 are limited to extraembryonic tissues. Indeed, placentas of the double-deficient animals showed decreased vascularization, and the ability of placental epithelium to establish a functional feto-maternal barrier was severely diminished. Interestingly, molecular analysis suggested that the barrier defect was associated with a selective deficiency in the expression of the tight junction protein, claudin-1. Our results reveal unexpected complementary roles of matriptase-prostasin- and PAR-2-dependent proteolytic signaling in the establishment of placental epithelial barrier function and overall embryonic survival. Development of mammalian embryos is dependent on an efficient exchange of nutrients, oxygen, and waste products between the mother and the embryo. The interface between the two systems is provided by the placenta in a form of a specialized epithelium that both facilitates the transport of molecules between the mother and the embryo and screens the substances that can pass between the maternal and fetal tissues. We now show that two independent signaling pathways that include the serine proteases, matriptase and prostasin, and a G protein-coupled receptor PAR-2, are critical for the establishment of a functional feto-maternal interface by specifically regulating the barrier properties of the placental epithelium. Because aberrant formation of epithelial barriers is an underlying feature of a great variety of human developmental abnormalities, the identification of the two protease-dependent signaling pathways critical for the barrier formation in embryonic tissues may help pinpoint molecular mechanisms involved in the etiology of these conditions.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Diane E. Peters
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
- Program of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Peter Kosa
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France
- Université Paris-Descartes, Paris, France
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
87
|
Fang JD, Lee SL. Matriptase is required for the active form of hepatocyte growth factor induced Met, focal adhesion kinase and protein kinase B activation on neural stem/progenitor cell motility. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1285-94. [DOI: 10.1016/j.bbamcr.2014.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/19/2014] [Accepted: 03/23/2014] [Indexed: 12/31/2022]
|
88
|
Friis S, Sales KU, Schafer JM, Vogel LK, Kataoka H, Bugge TH. The protease inhibitor HAI-2, but not HAI-1, regulates matriptase activation and shedding through prostasin. J Biol Chem 2014; 289:22319-32. [PMID: 24962579 DOI: 10.1074/jbc.m114.574400] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The membrane-anchored serine proteases, matriptase and prostasin, and the membrane-anchored serine protease inhibitors, hepatocyte growth factor activator inhibitor (HAI)-1 and HAI-2, are critical effectors of epithelial development and postnatal epithelial homeostasis. Matriptase and prostasin form a reciprocal zymogen activation complex that results in the formation of active matriptase and prostasin that are targets for inhibition by HAI-1 and HAI-2. Conflicting data, however, have accumulated as to the existence of auxiliary functions for both HAI-1 and HAI-2 in regulating the intracellular trafficking and activation of matriptase. In this study, we, therefore, used genetically engineered mice to determine the effect of ablation of endogenous HAI-1 and endogenous HAI-2 on endogenous matriptase expression, subcellular localization, and activation in polarized intestinal epithelial cells. Whereas ablation of HAI-1 did not affect matriptase in epithelial cells of the small or large intestine, ablation of HAI-2 resulted in the loss of matriptase from both tissues. Gene silencing studies in intestinal Caco-2 cell monolayers revealed that this loss of cell-associated matriptase was mechanistically linked to accelerated activation and shedding of the protease caused by loss of prostasin regulation by HAI-2. Taken together, these data indicate that HAI-1 regulates the activity of activated matriptase, whereas HAI-2 has an essential role in regulating prostasin-dependent matriptase zymogen activation.
Collapse
Affiliation(s)
- Stine Friis
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, and the Department of Cellular and Molecular Medicine, Faculty of Health Science, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Katiuchia Uzzun Sales
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, and Clinical Research Core, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Jeffrey Martin Schafer
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, and the College of Medicine, The Ohio State University, Columbus, Ohio 43210, and
| | - Lotte K Vogel
- the Department of Cellular and Molecular Medicine, Faculty of Health Science, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Hiroaki Kataoka
- the Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Thomas H Bugge
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, and
| |
Collapse
|
89
|
Pal K, Mukhopadhyay S. Primary cilium and sonic hedgehog signaling during neural tube patterning: Role of GPCRs and second messengers. Dev Neurobiol 2014; 75:337-48. [DOI: 10.1002/dneu.22193] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/09/2014] [Accepted: 05/22/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Kasturi Pal
- Department of Cell Biology; UT Southwestern Medical Center; Dallas Texas 75390
| | - Saikat Mukhopadhyay
- Department of Cell Biology; UT Southwestern Medical Center; Dallas Texas 75390
| |
Collapse
|
90
|
Peters DE, Szabo R, Friis S, Shylo NA, Uzzun Sales K, Holmbeck K, Bugge TH. The membrane-anchored serine protease prostasin (CAP1/PRSS8) supports epidermal development and postnatal homeostasis independent of its enzymatic activity. J Biol Chem 2014; 289:14740-9. [PMID: 24706745 DOI: 10.1074/jbc.m113.541318] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The membrane-anchored serine protease prostasin (CAP1/PRSS8) is part of a cell surface proteolytic cascade that is essential for epithelial barrier formation and homeostasis. Here, we report the surprising finding that prostasin executes these functions independent of its own enzymatic activity. Prostasin null (Prss8(-/-)) mice lack barrier formation and display fatal postnatal dehydration. In sharp contrast, mice homozygous for a point mutation in the Prss8 gene, which causes the substitution of the active site serine within the catalytic histidine-aspartate-serine triad with alanine and renders prostasin catalytically inactive (Prss8(Cat-/Cat-) mice), develop barrier function and are healthy when followed for up to 20 weeks. This striking difference could not be explained by genetic modifiers or by maternal effects, as these divergent phenotypes were displayed by Prss8(-/-) and Prss8(Cat-/Cat-) mice born within the same litter. Furthermore, Prss8(Cat-/Cat-) mice were able to regenerate epidermal covering following cutaneous wounding. This study provides the first demonstration that essential in vivo functions of prostasin are executed by a non-enzymatic activity of this unique membrane-anchored serine protease.
Collapse
Affiliation(s)
- Diane E Peters
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, the Program of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Roman Szabo
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Stine Friis
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, the Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Natalia A Shylo
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Katiuchia Uzzun Sales
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, the Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892
| | - Kenn Holmbeck
- the Connective Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Thomas H Bugge
- From the Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892,
| |
Collapse
|
91
|
Wang JK, Teng IJ, Lo TJ, Moore S, Yeo YH, Teng YC, Kaul M, Chen CC, Zuo AH, Chou FP, Yang X, Tseng IC, Johnson MD, Lin CY. Matriptase autoactivation is tightly regulated by the cellular chemical environments. PLoS One 2014; 9:e93899. [PMID: 24705933 PMCID: PMC3976350 DOI: 10.1371/journal.pone.0093899] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 03/07/2014] [Indexed: 12/17/2022] Open
Abstract
The ability of cells to rapidly detect and react to alterations in their chemical environment, such as pH, ionic strength and redox potential, is essential for cell function and survival. We present here evidence that cells can respond to such environmental alterations by rapid induction of matriptase autoactivation. Specifically, we show that matriptase autoactivation can occur spontaneously at physiological pH, and is significantly enhanced by acidic pH, both in a cell-free system and in living cells. The acid-accelerated autoactivation can be attenuated by chloride, a property that may be part of a safety mechanism to prevent unregulated matriptase autoactivation. Additionally, the thio-redox balance of the environment also modulates matriptase autoactivation. Using the cell-free system, we show that matriptase autoactivation is suppressed by cytosolic reductive factors, with this cytosolic suppression being reverted by the addition of oxidizing agents. In living cells, we observed rapid induction of matriptase autoactivation upon exposure to toxic metal ions known to induce oxidative stress, including CoCl2 and CdCl2. The metal-induced matriptase autoactivation is suppressed by N-acetylcysteine, supporting the putative role of altered cellular redox state in metal induced matriptase autoactivation. Furthermore, matriptase knockdown rendered cells more susceptible to CdCl2-induced cell death compared to control cells. This observation implies that the metal-induced matriptase autoactivation confers cells with the ability to survive exposure to toxic metals and/or oxidative stress. Our results suggest that matriptase can act as a cellular sensor of the chemical environment of the cell that allows the cell to respond to and protect itself from changes in the chemical milieu.
Collapse
Affiliation(s)
- Jehng-Kang Wang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - I-Jou Teng
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Jen Lo
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Sean Moore
- Geenebaum Cancer Center, University of Maryland, Baltimore, Maryland, United States of America
| | - Yee Hui Yeo
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yun-Chung Teng
- Department of Biomedical Engineering National Yang Ming University, Taipei, Taiwan
| | - Malvika Kaul
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Chiann-Chyi Chen
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States of America
| | - Annie Hong Zuo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, United States of America
| | - Fen-Pai Chou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, United States of America
| | - Xiaoyu Yang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, United States of America
| | - I-Chu Tseng
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, United States of America
| | - Michael D. Johnson
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, United States of America
| | - Chen-Yong Lin
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, United States of America
- * E-mail:
| |
Collapse
|
92
|
de Veer SJ, Furio L, Harris JM, Hovnanian A. Proteases: common culprits in human skin disorders. Trends Mol Med 2014; 20:166-78. [DOI: 10.1016/j.molmed.2013.11.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 11/24/2013] [Accepted: 11/25/2013] [Indexed: 12/17/2022]
|
93
|
Sales KU, Friis S, Konkel JE, Godiksen S, Hatakeyama M, Hansen KK, Rogatto SR, Szabo R, Vogel LK, Chen W, Gutkind JS, Bugge TH. Non-hematopoietic PAR-2 is essential for matriptase-driven pre-malignant progression and potentiation of ras-mediated squamous cell carcinogenesis. Oncogene 2014; 34:346-56. [PMID: 24469043 PMCID: PMC4112178 DOI: 10.1038/onc.2013.563] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/06/2013] [Accepted: 12/10/2013] [Indexed: 01/27/2023]
Abstract
The membrane-anchored serine protease, matriptase, is consistently dysregulated in a range of human carcinomas, and high matriptase activity correlates with poor prognosis. Furthermore, matriptase is unique among tumor-associated proteases in that epithelial stem cell expression of the protease suffices to induce malignant transformation. Here, we use genetic epistasis analysis to identify proteinase-activated receptor (PAR)-2-dependent inflammatory signaling as an essential component of matriptase-mediated oncogenesis. In cell-based assays, matriptase was a potent activator of PAR-2, and PAR-2 activation by matriptase caused robust induction of nuclear factor (NF)κB through Gαi. Importantly, genetic elimination of PAR-2 from mice completely prevented matriptase-induced pre-malignant progression, including inflammatory cytokine production, inflammatory cell recruitment, epidermal hyperplasia and dermal fibrosis. Selective ablation of PAR-2 from bone marrow-derived cells did not prevent matriptase-driven pre-malignant progression, indicating that matriptase activates keratinocyte stem cell PAR-2 to elicit its pro-inflammatory and pro-tumorigenic effects. When combined with previous studies, our data suggest that dual induction of PAR-2-NFκB inflammatory signaling and PI3K-Akt-mTor survival/proliferative signaling underlies the transforming potential of matriptase and may contribute to pro-tumorigenic signaling in human epithelial carcinogenesis.
Collapse
Affiliation(s)
- K U Sales
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S Friis
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - J E Konkel
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S Godiksen
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark [3] Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - M Hatakeyama
- 1] Department of Urology, Faculty of Medicine, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil [2] AC Camargo Cancer Center, Sao Paulo, Brazil
| | - K K Hansen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S R Rogatto
- 1] Department of Urology, Faculty of Medicine, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil [2] AC Camargo Cancer Center, Sao Paulo, Brazil
| | - R Szabo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - L K Vogel
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - W Chen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - J S Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - T H Bugge
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
94
|
Ye J, Kawaguchi M, Haruyama Y, Kanemaru A, Fukushima T, Yamamoto K, Lin CY, Kataoka H. Loss of hepatocyte growth factor activator inhibitor type 1 participates in metastatic spreading of human pancreatic cancer cells in a mouse orthotopic transplantation model. Cancer Sci 2013; 105:44-51. [PMID: 24147538 PMCID: PMC4317873 DOI: 10.1111/cas.12306] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 10/09/2013] [Accepted: 10/18/2013] [Indexed: 12/18/2022] Open
Abstract
Hepatocyte growth factor activator inhibitor type 1 (HAI-1) is a membrane-bound serine protease inhibitor that is expressed on the surface of epithelial and carcinoma cells. On the cell surface, HAI-1 regulates membrane-anchored serine proteases, with matriptase being the most critical target. Matriptase is involved in pericellular processing of biologically active molecules, including protease-activated receptor-2 (PAR-2). Previously we reported that S2-CP8 cells, a metastatic variant of the SUIT-2 human pancreatic adenocarcinoma cell line, showed markedly decreased HAI-1 expression. To assess the significance of HAI-1 loss in invasion and spontaneous metastasis of S2-CP8 cells, we established stable S2-CP8 sublines that expressed HAI-1 under the control of a tetracycline-regulated promoter. In vitro migration and invasion assays revealed inhibitory effects of HAI-1 on S2-CP8 cell migration and invasion. Matriptase activity was suppressed by the expression of HAI-1. As the enhanced invasiveness in the absence of HAI-1 was alleviated by knockdown of matriptase by 81% and of PAR-2 completely, and PAR-2 antagonist also suppressed the invasion, matriptase-mediated PAR-2 activation is involved in HAI-1 loss-induced invasion of S2-CP8 cells. We then analyzed the effect of HAI-1 expression on metastasis of S2-CP8 cells in vivo using a nude mouse orthotopic xenograft model. Although approximately 50% of the control mice developed distant metastasis, mice treated with doxycycline to induce HAI-1 expression did not develop metastasis. These data indicate that HAI-1 loss contributes to invasion and dissemination of a highly metastatic subline of SUIT-2, suggesting crucial roles for the balance of pericellular serine proteases/inhibitors in pancreatic cancer progression.
Collapse
Affiliation(s)
- Jingjia Ye
- Section of Oncopathology and Regenerative Biology, Department of Pathology, University of Miyazaki, Miyazaki, Japan; Clinical Research Center, The 2nd Affiliated Hospital School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Godiksen S, Soendergaard C, Friis S, Jensen JK, Bornholdt J, Sales KU, Huang M, Bugge TH, Vogel LK. Detection of active matriptase using a biotinylated chloromethyl ketone peptide. PLoS One 2013; 8:e77146. [PMID: 24204759 PMCID: PMC3799725 DOI: 10.1371/journal.pone.0077146] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/22/2013] [Indexed: 01/20/2023] Open
Abstract
Matriptase is a member of the family of type II transmembrane serine proteases that is essential for development and maintenance of several epithelial tissues. Matriptase is synthesized as a single-chain zymogen precursor that is processed into a two-chain disulfide-linked form dependent on its own catalytic activity leading to the hypothesis that matriptase functions at the pinnacle of several protease induced signal cascades. Matriptase is usually found in either its zymogen form or in a complex with its cognate inhibitor hepatocyte growth factor activator inhibitor 1 (HAI-1), whereas the active non-inhibited form has been difficult to detect. In this study, we have developed an assay to detect enzymatically active non-inhibitor-complexed matriptase by using a biotinylated peptide substrate-based chloromethyl ketone (CMK) inhibitor. Covalently CMK peptide-bound matriptase is detected by streptavidin pull-down and subsequent analysis by Western blotting. This study presents a novel assay for detection of enzymatically active matriptase in living human and murine cells. The assay can be applied to a variety of cell systems and species.
Collapse
Affiliation(s)
- Sine Godiksen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Proteases and Tissue Remodeling Unit, National Institute of Dental and Craniofacial Research, Bethesda, Maryland, United States of America
| | | | - Stine Friis
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Proteases and Tissue Remodeling Unit, National Institute of Dental and Craniofacial Research, Bethesda, Maryland, United States of America
| | - Jan K. Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Danish-Chinese Centre for Proteases and Cancer
| | - Jette Bornholdt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Katiuchia Uzzun Sales
- Proteases and Tissue Remodeling Unit, National Institute of Dental and Craniofacial Research, Bethesda, Maryland, United States of America
| | - Mingdong Huang
- Danish-Chinese Centre for Proteases and Cancer
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Fuzhou, Fujian, China
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Unit, National Institute of Dental and Craniofacial Research, Bethesda, Maryland, United States of America
| | - Lotte K. Vogel
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
96
|
Abstract
The generation of knock out mice urged researchers, not always voluntarily, to newly define developmental functions of the gene knocked out. Among others, this has led to the establishment of the field of developmental haemostasis. The work in this field identified a role of coagulation proteases and their regulators independent of haemostasis in the embryo proper. Rather, coagulation proteases regulate cellular function through receptor dependent signalling in the embryo proper, both within and outside the vasculature. Likewise, coagulation proteases modulate placental development independent of haemostasis through mechanisms involving the activation of maternal myeloid derived cells. The following review summarizes the current knowledge in the field of developmental haemostasis and pinpoints open questions within this evolving field.
Collapse
Affiliation(s)
- Muhammed Kashif
- Department of Internal Medicine I and Clinical Chemistry, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | | |
Collapse
|
97
|
Lin H, Liu AP, Smith TH, Trejo J. Cofactoring and dimerization of proteinase-activated receptors. Pharmacol Rev 2013; 65:1198-213. [PMID: 24064459 PMCID: PMC3799237 DOI: 10.1124/pr.111.004747] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Proteinase-activated receptors (PARs) are G protein-coupled receptors that transmit cellular responses to extracellular proteases and have important functions in vascular physiology, development, inflammation, and cancer progression. The established paradigm for PAR activation involves proteolytic cleavage of the extracellular N terminus, which reveals a new N terminus that functions as a tethered ligand by binding intramolecularly to the receptor to trigger transmembrane signaling. Most cells express more than one PAR, which can influence the mode of PAR activation and signaling. Clear examples include murine PAR3 cofactoring of PAR4 and transactivation of PAR2 by PAR1. Thrombin binds to and cleaves murine PAR3, which facilitates PAR4 cleavage and activation. This process is essential for thrombin signaling and platelet activation, since murine PAR3 cannot signal alone. Although PAR1 and PAR4 are both competent to signal, PAR1 is able to act as a cofactor for PAR4, facilitating more rapid cleavage and activation by thrombin. PAR1 can also facilitate PAR2 activation through a different mechanism. Cleavage of the PAR1 N terminus by thrombin generates a tethered ligand domain that can bind intermolecularly to PAR2 to activate signaling. Thus, PARs can regulate each other's activity by localizing thrombin when in complex with PAR3 and PAR4 or by cleaved PAR1, providing its tethered ligand domain for PAR2 activation. The ability of PARs to cofactor or transactivate other PARs would necessitate that the two receptors be in close proximity, likely in the form of a heterodimer. Here, we discuss the cofactoring and dimerization of PARs and the functional consequences on signaling.
Collapse
Affiliation(s)
- Huilan Lin
- University of California, San Diego, 9500 Gilman Drive, Biomedical Sciences Building, MC-0636, La Jolla, CA 92093.
| | | | | | | |
Collapse
|
98
|
Singh CK, Kumar A, Lavoie HA, Dipette DJ, Singh US. Diabetic complications in pregnancy: is resveratrol a solution? Exp Biol Med (Maywood) 2013; 238:482-90. [PMID: 23436883 DOI: 10.1177/1535370212473704] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Diabetes is a metabolic disorder that, during pregnancy, may affect fetal development. Fetal outcome depends on the type of diabetes present, the concentration of blood glucose and the extent of fetal exposure to elevated or frequently fluctuating glucose concentrations. The result of some diabetic pregnancies will be embryonic developmental abnormalities, a condition referred to as diabetic embryopathy. Tight glycemic control in type 1 diabetes during pregnancy using insulin therapy together with folic acid supplementation are partially able to prevent diabetic embryopathy; however, the protection is not complete and additional interventions are needed. Resveratrol, a polyphenol found largely in the skins of red grapes, is known to have antidiabetic action and is in clinical trials for the treatment of diabetes, insulin resistance, obesity and metabolic syndrome. Studies of resveratrol in a rodent model of diabetic embryopathy reveal that it significantly improves the embryonic outcome in terms of diminishing developmental abnormalities. Improvements in maternal and embryonic outcomes observed in rodent models may arise from resveratrol's antioxidative potential, antidiabetic action and antidyslipidemic nature. Whether resveratrol will have similar actions in human diabetic pregnancy is unknown. Here, we review the potential therapeutic use of resveratrol in diabetes and diabetic pregnancy.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Pathology, School of Medicine, 6311 Garners Ferry Road, Columbia, SC 29209, USA
| | | | | | | | | |
Collapse
|
99
|
Singh CK, Kumar A, Lavoie HA, Dipette DJ, Singh US. Diabetic complications in pregnancy: is resveratrol a solution? EXPERIMENTAL BIOLOGY AND MEDICINE (MAYWOOD, N.J.) 2013. [PMID: 23436883 DOI: 10.1177/1535370212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Diabetes is a metabolic disorder that, during pregnancy, may affect fetal development. Fetal outcome depends on the type of diabetes present, the concentration of blood glucose and the extent of fetal exposure to elevated or frequently fluctuating glucose concentrations. The result of some diabetic pregnancies will be embryonic developmental abnormalities, a condition referred to as diabetic embryopathy. Tight glycemic control in type 1 diabetes during pregnancy using insulin therapy together with folic acid supplementation are partially able to prevent diabetic embryopathy; however, the protection is not complete and additional interventions are needed. Resveratrol, a polyphenol found largely in the skins of red grapes, is known to have antidiabetic action and is in clinical trials for the treatment of diabetes, insulin resistance, obesity and metabolic syndrome. Studies of resveratrol in a rodent model of diabetic embryopathy reveal that it significantly improves the embryonic outcome in terms of diminishing developmental abnormalities. Improvements in maternal and embryonic outcomes observed in rodent models may arise from resveratrol's antioxidative potential, antidiabetic action and antidyslipidemic nature. Whether resveratrol will have similar actions in human diabetic pregnancy is unknown. Here, we review the potential therapeutic use of resveratrol in diabetes and diabetic pregnancy.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Pathology, School of Medicine, 6311 Garners Ferry Road, Columbia, SC 29209, USA
| | | | | | | | | |
Collapse
|
100
|
Massarwa R, Ray HJ, Niswander L. Morphogenetic movements in the neural plate and neural tube: mouse. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 3:59-68. [DOI: 10.1002/wdev.120] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- R'ada Massarwa
- Department of Molecular Genetics; The Weizmann Institute of Science; Rehovot Israel
| | - Heather J. Ray
- Department of Pediatrics, Cell Biology Stem Cells and Development Graduate Program; University of Colorado School of Medicine and Children's Hospital Colorado; Aurora CO USA
| | - Lee Niswander
- Department of Pediatrics, Cell Biology Stem Cells and Development Graduate Program; University of Colorado School of Medicine and Children's Hospital Colorado; Aurora CO USA
| |
Collapse
|