1
|
Kojima K, Katsuno T, Kishimoto Y, Mizuta M, Nakamura R, Ohnishi H, Yamada K, Kawai Y, Tateya I, Omori K. In vitro model to evaluate effect of acidic pepsin on vocal fold barrier function. Biochem Biophys Res Commun 2024; 732:150401. [PMID: 39033554 DOI: 10.1016/j.bbrc.2024.150401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
The pathophysiology of laryngopharyngeal reflux (LPR) and its impact on the vocal fold is not well understood, but may involve acid damage to vocal fold barrier functions. Two different components encompass vocal fold barrier function: the mucus barrier and tight junctions. Mucus retained on epithelial microprojections protects the inside of the vocal fold by neutralizing acidic damage. Tight junctions control permeability between cells. Here we developed an in vitro experimental system to evaluate acidic injury and repair of vocal fold barrier functions. We first established an in vitro model of rat vocal fold epithelium that could survive at least one week after barrier function maturation. The model enabled repeated evaluation of the course of vocal fold repair processes. Then, an injury experiment was conducted in which vocal fold cells were exposed to a 5-min treatment with acidic pepsin that injured tight junctions and cell surface microprojections. Both of them healed within one day of injury. Comparing vocal fold cells treated with acid alone with cells treated with acidic pepsin showed that acidic pepsin had a stronger effect on intercellular permeability than acid alone, whereas pepsin had little effect on microprojections. This result suggests that the proteolytic action of pepsin has a larger effect on protein-based tight junctions than on phospholipids in microprojections. This experimental system could contribute to a better understanding of vocal fold repair processes after chemical or physical injuries, as well as voice problems due to LPR pathogenesis.
Collapse
Affiliation(s)
| | - Tatsuya Katsuno
- Center of Anatomical, Pathological and Forensic Medical Researches, Graduate School of Medicine, Kyoto University, Japan
| | - Yo Kishimoto
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Japan.
| | | | - Ryosuke Nakamura
- Department of Rehabilitation Medicine, New York University Grossman School of Medicine, New York, USA
| | - Hiroe Ohnishi
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Koichiro Yamada
- Department of Otolaryngology Head and Neck Surgery, Kurashiki Central Hospital, Japan
| | - Yoshitaka Kawai
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Ichiro Tateya
- Department of Otorhinolaryngology, Fujita Health University, Japan
| | - Koichi Omori
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Japan
| |
Collapse
|
2
|
Monaco A, Axis J, Amsler K. Simple graphical approach to investigate differences in transepithelial paracellular leak pathway permeability. Physiol Rep 2022; 10:e15202. [PMID: 35274827 PMCID: PMC8915387 DOI: 10.14814/phy2.15202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023] Open
Abstract
Although many studies have reported differences in epithelial paracellular Leak Pathway permeability following genetic manipulations and treatment with various agents, the basis for these differences remains mostly unclear. Two primary mechanisms which could underlie differences in Leak Pathway permeability are differences in the density of Leak Pathway openings and differences in the opening size. Using a computational approach, we demonstrate that these two possibilities can be readily distinguished graphically by comparing the apparent paracellular permeabilities of a size panel of solutes measured across different cell layers. Using this approach, we demonstrated that depletion of ZO-1 protein in MDCK Type II renal epithelial cells decreased Leak Pathway opening size and increased opening density. Depletion of ZO-2 protein either had no effect or minimally decreased opening size and did not markedly change opening density. Comparison of MDCK Type II cells with MDCK Type I cells revealed that Type I cells exhibited a substantially smaller Leak Pathway permeability than did Type II cells. This lower permeability was due to a decrease in opening density with little or no change in opening size. These results demonstrate the utility of this approach to provide insights into the basis for observed differences in epithelial Leak Pathway permeability. This approach has wide applications including analysis of the molecular basis for Leak Pathway permeability, the effects of specific manipulations on Leak Pathway permeability properties, and the effects of permeation enhancers on Leak Pathway permeability properties.
Collapse
Affiliation(s)
- Ashley Monaco
- Department of Biomedical SciencesNYIT College of Osteopathic MedicineOld WestburyNew YorkUSA
| | - Josephine Axis
- Department of Biomedical SciencesNYIT College of Osteopathic MedicineOld WestburyNew YorkUSA
| | - Kurt Amsler
- Department of Biomedical SciencesNYIT College of Osteopathic MedicineOld WestburyNew YorkUSA
| |
Collapse
|
3
|
Plaunt AJ, Nguyen TL, Corboz MR, Malinin VS, Cipolla DC. Strategies to Overcome Biological Barriers Associated with Pulmonary Drug Delivery. Pharmaceutics 2022; 14:302. [PMID: 35214039 PMCID: PMC8880668 DOI: 10.3390/pharmaceutics14020302] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 02/01/2023] Open
Abstract
While the inhalation route has been used for millennia for pharmacologic effect, the biological barriers to treating lung disease created real challenges for the pharmaceutical industry until sophisticated device and formulation technologies emerged over the past fifty years. There are now several inhaled device technologies that enable delivery of therapeutics at high efficiency to the lung and avoid excessive deposition in the oropharyngeal region. Chemistry and formulation technologies have also emerged to prolong retention of drug at the active site by overcoming degradation and clearance mechanisms, or by reducing the rate of systemic absorption. These technologies have also been utilized to improve tolerability or to facilitate uptake within cells when there are intracellular targets. This paper describes the biological barriers and provides recent examples utilizing formulation technologies or drug chemistry modifications to overcome those barriers.
Collapse
Affiliation(s)
- Adam J. Plaunt
- Insmed Incorporated, Bridgewater, NJ 08807, USA; (T.L.N.); (M.R.C.); (V.S.M.); (D.C.C.)
| | | | | | | | | |
Collapse
|
4
|
Ravikrishnan A, Fowler EW, Stuffer AJ, Jia X. Hydrogel-Supported, Engineered Model of Vocal Fold Epithelium. ACS Biomater Sci Eng 2021; 7:4305-4317. [PMID: 33635635 DOI: 10.1021/acsbiomaterials.0c01741] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is a critical need for the establishment of an engineered model of the vocal fold epithelium that can be used to gain understanding of its role in vocal fold health, disease, and facilitate the development of new treatment options. Toward this goal, we isolated primary vocal fold epithelial cells (VFECs) from healthy porcine larynxes and used them within passage 3. Culture-expanded VFECs expressed the suprabasal epithelial marker cytokeratin 13 and intercellular junctional proteins occludin, E-cadherin, and zonula occludens-1. To establish the engineered model, we cultured VFECs on a hyaluronic acid-derived synthetic basement membrane displaying fibronectin-derived integrin-binding peptide (RGDSP) and/or laminin 111-derived syndecan-binding peptide AG73 (RKRLQVQLSIRT). Our results show that matrix stiffness and composition cooperatively regulate the adhesion, proliferation, and stratification of VFECs. Cells cultured on hydrogels with physiological stiffness (elastic shear modulus, G' = 1828 Pa) adopted a cobblestone morphology with close cell-cell contacts, whereas those on softer matrices (G' = 41 Pa) were spindle shaped with extensive intracellular stress fibers. The development of stratified epithelium with proliferating basal cells and additional (1-2) suprabasal layers requires the presence of both RGDSP and AG73 peptide signals. Supplementation of cytokines produced by vimentin positive primary porcine vocal fold fibroblasts in the VFEC culture led to the establishment of 4-5 distinct cell layers. The engineered vocal fold epithelium resembled native tissue morphologically; expressed cytokeratin 13, mucin 1, and tight/adherens junction markers; and secreted basement membrane proteins collagen IV and laminin 5. Collectively, our results demonstrate that stiffness matching, cell-matrix engagement, and paracrine signaling cooperatively contribute to the stratification of VFECs. The engineered epithelium can be used as a versatile tool for investigations of genetic and molecular mechanisms in vocal fold health and disease.
Collapse
Affiliation(s)
- Anitha Ravikrishnan
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Eric W Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Alexander J Stuffer
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States.,Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States.,Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States.,Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19711, United States
| |
Collapse
|
5
|
Monaco A, Ovryn B, Axis J, Amsler K. The Epithelial Cell Leak Pathway. Int J Mol Sci 2021; 22:ijms22147677. [PMID: 34299297 PMCID: PMC8305272 DOI: 10.3390/ijms22147677] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
The epithelial cell tight junction structure is the site of the transepithelial movement of solutes and water between epithelial cells (paracellular permeability). Paracellular permeability can be divided into two distinct pathways, the Pore Pathway mediating the movement of small ions and solutes and the Leak Pathway mediating the movement of large solutes. Claudin proteins form the basic paracellular permeability barrier and mediate the movement of small ions and solutes via the Pore Pathway. The Leak Pathway remains less understood. Several proteins have been implicated in mediating the Leak Pathway, including occludin, ZO proteins, tricellulin, and actin filaments, but the proteins comprising the Leak Pathway remain unresolved. Many aspects of the Leak Pathway, such as its molecular mechanism, its properties, and its regulation, remain controversial. In this review, we provide a historical background to the evolution of the Leak Pathway concept from the initial examinations of paracellular permeability. We then discuss current information about the properties of the Leak Pathway and present current theories for the Leak Pathway. Finally, we discuss some recent research suggesting a possible molecular basis for the Leak Pathway.
Collapse
Affiliation(s)
- Ashley Monaco
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
| | - Ben Ovryn
- Department of Physics, New York Institute of Technology, Northern Boulevard, Old Westbury, NY 11568, USA;
| | - Josephine Axis
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
| | - Kurt Amsler
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
- Correspondence: ; Tel.: +1-516-686-3716
| |
Collapse
|
6
|
Hartmann C, Schwietzer YA, Otani T, Furuse M, Ebnet K. Physiological functions of junctional adhesion molecules (JAMs) in tight junctions. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183299. [DOI: 10.1016/j.bbamem.2020.183299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/24/2022]
|
7
|
Sassi A, Wang Y, Chassot A, Komarynets O, Roth I, Olivier V, Crambert G, Dizin E, Boscardin E, Hummler E, Feraille E. Interaction between Epithelial Sodium Channel γ-Subunit and Claudin-8 Modulates Paracellular Sodium Permeability in Renal Collecting Duct. J Am Soc Nephrol 2020; 31:1009-1023. [PMID: 32245797 DOI: 10.1681/asn.2019080790] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/15/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Water and solute transport across epithelia can occur via the transcellular or paracellular pathways. Tight junctions play a key role in mediating paracellular ion reabsorption in the kidney. In the renal collecting duct, which is a typical absorptive tight epithelium, coordination between transcellular sodium reabsorption and paracellular permeability may prevent the backflow of reabsorbed sodium to the tubular lumen along a steep electrochemical gradient. METHODS To investigate whether transcellular sodium transport controls tight-junction composition and paracellular permeability via modulating expression of the transmembrane protein claudin-8, we used cultured mouse cortical collecting duct cells to see how overexpression or silencing of epithelial sodium channel (ENaC) subunits and claudin-8 affect paracellular permeability. We also used conditional kidney tubule-specific knockout mice lacking ENaC subunits to assess the ENaC's effect on claudin-8 expression. RESULTS Overexpression or silencing of the ENaC γ-subunit was associated with parallel and specific changes in claudin-8 abundance. Increased claudin-8 abundance was associated with a reduction in paracellular permeability to sodium, whereas decreased claudin-8 abundance was associated with the opposite effect. Claudin-8 overexpression and silencing reproduced these functional effects on paracellular ion permeability. Conditional kidney tubule-specific ENaC γ-subunit knockout mice displayed decreased claudin-8 expression, confirming the cell culture experiments' findings. Importantly, ENaC β-subunit or α-subunit silencing or kidney tubule-specific β-ENaC or α-ENaC knockout mice did not alter claudin-8 abundance. CONCLUSIONS Our data reveal the specific coupling between ENaC γ-subunit and claudin-8 expression. This coupling may play an important role in preventing the backflow of reabsorbed solutes and water to the tubular lumen, as well as in coupling paracellular and transcellular sodium permeability.
Collapse
Affiliation(s)
- Ali Sassi
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Yubao Wang
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Alexandra Chassot
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Olga Komarynets
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland
| | - Isabelle Roth
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland
| | - Valérie Olivier
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Gilles Crambert
- Sorbonne University, Unité Mixte de Recherche (UMR) S1138, Cordeliers Research Center, Paris, France
| | - Eva Dizin
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Emilie Boscardin
- National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Edith Hummler
- National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Eric Feraille
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland .,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| |
Collapse
|
8
|
Hu M, Zhou T, Dezzutti CS, Rohan LC. The Effect of Commonly Used Excipients on the Epithelial Integrity of Human Cervicovaginal Tissue. AIDS Res Hum Retroviruses 2017; 32:992-1004. [PMID: 27611224 DOI: 10.1089/aid.2016.0014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pharmaceutical excipients are widely used in vaginal drug products. The epithelial integrity of the cervicovaginal tissue is important for HIV-1 prevention. However, the effects of excipients on cervicovaginal epithelium remain unknown. This study aims at assessing the effects of vaginal product excipients on the integrity of human cervicovaginal epithelium and on a lead HIV prevention antiretroviral drug, tenofovir (TFV). In the current study, nine excipients commonly used in vaginal formulations were incubated for 6 h with excised human ectocervical tissue. The effects of the excipients were examined by measuring the transepithelial electrical resistance (TEER), epithelial morphology, paracellular/transcellular permeability, and cell viability. The efficacy of TFV for preventing HIV-1 infection in the ex vivo cultured ectocervix was also tested. We found that disodium ethyl-enediaminetetraacetate (EDTA), sorbic acid, and benzoic acid had no effect on the tissue TEER. Butylated hydroxyanisole, glycerin, propylene glycol, methylparaben, and propylparaben slightly to moderately decreased tissue TEER, whereas citric acid significantly decreased the TEER in a time-dependent manner. Tissue morphology observed post-exposure strongly correlated with TEER data; however, a less strong correlation was observed between paracellular permeability and TEER data after exposure to different excipients. In addition, treatment with EDTA, methylparaben, and propylene glycol at tested levels had no effect on the efficacy of TFV in preventing tissue HIV-1 infection. In conclusion, the combined measurements of TEER, morphology, permeability, and viability using human cervicovaginal tissue represent a clinically relevant platform for safety evaluation of excipients and formulated products for HIV-1 prevention.
Collapse
Affiliation(s)
- Minlu Hu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Tian Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Charlene S. Dezzutti
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lisa C. Rohan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Mizuta M, Kurita T, Kimball EE, Rousseau B. Structurally and functionally characterized in vitro model of rabbit vocal fold epithelium. Tissue Cell 2017; 49:427-434. [PMID: 28395914 PMCID: PMC5467526 DOI: 10.1016/j.tice.2017.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/19/2022]
Abstract
In this paper, we describe a method for primary culture of a well differentiated electrically tight rabbit vocal fold epithelial cell multilayer and the measurement of transepithelial electrical resistance (TEER) for the evaluation of epithelial barrier function in vitro. Rabbit larynges were harvested and enzymatically treated to isolate vocal fold epithelial cells and to establish primary culture. Vocal fold epithelial cells were co-cultured with mitomycin C-treated feeder cells on collagen-coated plates. After 10-14 days in primary culture, cells were passaged and cultured until they achieved 70-90% confluence on collagen-coated plates. Epithelial cells were then passaged onto collagen-coated cell culture inserts using 4.5cm2 membrane filters (1.0μm pore size) with 10% fetal bovine serum or 30μg/mL bovine pituitary extract to investigate the effects of growth-promoting additives on TEER. Additional experiments were performed to investigate optimal seeding density (1.1, 2.2, 4.4, or 8.9×105 cells/cm2), the effect of co-culture with feeder cells, and the effect of passage number on epithelial barrier function. Characterization of in vitro cultures was performed using hematoxylin and eosin staining and immunostaining for vocal fold epithelial cell markers and tight junctions. Results revealed higher TEER in cells supplemented with fetal bovine serum compared to bovine pituitary extract. TEER was highest in cells passaged at a seeding density of 2.2×104 cells/cm2, and TEER was higher in cells at passage two than passage three. Ultrastructural experiments revealed a well-differentiated epithelial cell multilayer, expressing the epithelial cell markers CK13, CK14 and the tight junction proteins occludin and ZO-1.
Collapse
Affiliation(s)
- Masanobu Mizuta
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takashi Kurita
- Department of Otolaryngology, Vanderbilt University Medical Center, 1215 21st Avenue South, Medical Center East, Nashville, TN 37232, USA
| | - Emily E Kimball
- Department of Hearing and Speech Sciences, Vanderbilt University School of Medicine, 1215 21st Avenue South, Medical Center East, Nashville, TN 37232, USA
| | - Bernard Rousseau
- Department of Otolaryngology, Vanderbilt University Medical Center, and Hearing and Speech Sciences and Mechanical Engineering, Vanderbilt University School of Medicine, Medical Center East, 1215 21st Avenue South, Nashville, TN 37232, USA.
| |
Collapse
|
10
|
Abstract
Signaling via the Rho GTPases provides crucial regulation of numerous cell polarization events, including apicobasal (AB) polarity, polarized cell migration, polarized cell division and neuronal polarity. Here we review the relationships between the Rho family GTPases and epithelial AB polarization events, focusing on the 3 best-characterized members: Rho, Rac and Cdc42. We discuss a multitude of processes that are important for AB polarization, including lumen formation, apical membrane specification, cell-cell junction assembly and maintenance, as well as tissue polarity. Our discussions aim to highlight the immensely complex regulatory mechanisms that encompass Rho GTPase signaling during AB polarization. More specifically, in this review we discuss several emerging common themes, that include: 1) the need for Rho GTPase activities to be carefully balanced in both a spatial and temporal manner through a multitude of mechanisms; 2) the existence of signaling feedback loops and crosstalk to create robust cellular responses; and 3) the frequent multifunctionality that exists among AB polarity regulators. Regarding this latter theme, we provide further discussion of the potential plasticity of the cell polarity machinery and as a result the possible implications for human disease.
Collapse
Key Words
- AB, Apicobasal
- AJ, Adherens junction
- Amot, Angiomotin
- Arp2/3, Actin-related protein-2/3
- Baz, Bazooka
- C. elegans, Caenorhabditis elegans
- CA, Constitutively-active
- CD2AP, CD2-associated protein
- Caco2, Human colon carcinoma
- Cdc42
- Cora, Coracle
- Crb, Crumbs
- DN, Dominant-negative
- Dia1, Diaphanous-related formin 1
- Dlg, Discs large
- Drosophila, Drosophila melanogaster
- Dys-β, Dystrobrevin-β
- ECM, Extracellular matrix
- Ect2, Epithelial cell transforming sequence 2 oncogene
- Eya1, Eyes absent 1
- F-actin, Filamentous actin
- FRET, Fluorescence resonance energy transfer
- GAP, GTPase-activating protein
- GDI, Guanine nucleotide dissociation inhibitor
- GEF, Guanine nucleotide exchange factor
- GTPases
- JACOP, Junction-associated coiled-coiled protein
- JAM, Junctional adhesion molecule
- LKB1, Liver kinase B1
- Lgl, Lethal giant larvae
- MDCK, Madin-Darby canine kidney
- MTOC, Microtubule-organizing center
- NrxIV, Neurexin IV
- Pals1, Protein associated with Lin-7 1
- Par, Partitioning-defective
- Patj, Pals1-associated TJ protein
- ROCK, Rho-associated kinase
- Rac
- Rho
- Rich1, RhoGAP interacting with CIP4 homologues
- S. cerevisiae, Saccharomyces cerevisiae
- S. pombe, Schizosaccharomyces pombe
- SH3BP1, SH3-domain binding protein 1
- Scrib, Scribble
- Std, Stardust
- TEM4, Tumor endothelial marker 4
- TJ, Tight junction
- Tiam1, T-cell lymphoma invasion and metastasis-inducing protein 1
- WASp, Wiskott-aldrich syndrome protein
- Yrt, Yurt
- ZA, zonula adherens
- ZO, Zonula occludens
- aPKC, Atypical Protein Kinase C
- apicobasal
- epithelia
- junction
- par
- polarity
- α-cat, Alpha-catenin
- β-cat, Beta-Catenin
- β2-syn, Beta-2-syntrophin
Collapse
Affiliation(s)
- Natalie Ann Mack
- a School of Life Sciences; Queens Medical Center ; University of Nottingham ; Nottingham , UK
| | | |
Collapse
|
11
|
Levendoski EE, Leydon C, Thibeault SL. Vocal fold epithelial barrier in health and injury: a research review. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2014; 57:1679-91. [PMID: 24686981 PMCID: PMC4557797 DOI: 10.1044/2014_jslhr-s-13-0283] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 03/04/2014] [Indexed: 05/07/2023]
Abstract
PURPOSE Vocal fold epithelium is composed of layers of individual epithelial cells joined by junctional complexes constituting a unique interface with the external environment. This barrier provides structural stability to the vocal folds and protects underlying connective tissue from injury while being nearly continuously exposed to potentially hazardous insults, including environmental or systemic-based irritants such as pollutants and reflux, surgical procedures, and vibratory trauma. Small disruptions in the epithelial barrier may have a large impact on susceptibility to injury and overall vocal health. The purpose of this article is to provide a broad-based review of current knowledge of the vocal fold epithelial barrier. METHOD A comprehensive review of the literature was conducted. Details of the structure of the vocal fold epithelial barrier are presented and evaluated in the context of function in injury and pathology. The importance of the epithelial-associated vocal fold mucus barrier is also introduced. RESULTS/CONCLUSIONS Information presented in this review is valuable for clinicians and researchers as it highlights the importance of this understudied portion of the vocal folds to overall vocal health and disease. Prevention and treatment of injury to the epithelial barrier is a significant area awaiting further investigation.
Collapse
|
12
|
Martin-Martin N, Dan Q, Amoozadeh Y, Waheed F, McMorrow T, Ryan MP, Szászi K. RhoA and Rho kinase mediate cyclosporine A and sirolimus-induced barrier tightening in renal proximal tubular cells. Int J Biochem Cell Biol 2011; 44:178-88. [PMID: 22062948 DOI: 10.1016/j.biocel.2011.10.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 09/29/2011] [Accepted: 10/24/2011] [Indexed: 10/15/2022]
Abstract
The regulation and maintenance of the paracellular transport in renal tubular epithelia is vital for kidney functions. Combination of the immunosuppressant drugs cyclosporine A (CsA) and sirolimus (SRL) exerts powerful immunosuppression, but also causes nephrotoxicity. We have previously shown that CsA and SRL elevate transepithelial resistance (TER) in kidney tubular cells partly through MEK/ERK1/2. In this work we examined the hypothesis that the RhoA pathway may also be mediating effects of CsA and SRL. We show that CsA and the CsA/SRL combination activated RhoA, induced cofilin phosphorylation and promoted stress fiber generation. The Rho kinase (ROK) inhibitor, Y27632, prevented CsA and CsA/SRL-induced cofilin phosphorylation and actin remodelling, reduced the TER increase and prevented the rise in claudin-7 levels caused by the drugs. Expression of the exchange factor GEF-H1/lfc was elevated in cells treated with CsA and CsA/SRL. GEF-H1 silencing inhibited RhoA activation by ≈50%, and potently reduced cofilin phosphorylation and stress fiber formation induced by CsA and CsA/SRL. However, GEF-H1 downregulation did not prevent the TER change. Thus the Rho/Rho kinase pathway was involved in mediating CsA and CsA/SRL-induced cytoskeleton rearrangement and TER changes via claudin-7 expression. Our data however point to differential regulation of Rho activation involved in central cytoskeleton remodelling, that is GEF-H1-dependent and junctional permeability that does not require GEF-H1.
Collapse
Affiliation(s)
- Natalia Martin-Martin
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | | | |
Collapse
|
13
|
Wölke S, Ackermann N, Heesemann J. The Yersinia enterocolitica type 3 secretion system (T3SS) as toolbox for studying the cell biological effects of bacterial Rho GTPase modulating T3SS effector proteins. Cell Microbiol 2011; 13:1339-57. [PMID: 21718421 DOI: 10.1111/j.1462-5822.2011.01623.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The bacterial effector proteins IpgB(1) and IpgB(2) of Shigella and Map of Escherichia coli activate the Rho GTPases Rac1, RhoA and Cdc42, respectively, whereas YopE and YopT of Yersinia inhibit these Rho family GTPases. We established a Yersinia toolbox which allows to study the cellular effects of these effectors in different combinations in the context of Yersinia type 3 secretion system (Ysc)-T3SS-mediated injection into HeLa cells. For this purpose hybrid proteins were constructed by fusion of YopE with the effector protein of interest. As expected, injected hybrid proteins induced membrane ruffles and Yersinia uptake for IpgB(1) , stress fibres for IpgB(2) and microspikes for Map. By co-infection experiments we could demonstrate (i) IpgB(2) -mediated and ROCK-dependent inhibition of IpgB(1) -mediated Rac1 effects, (ii) YopT-mediated suppression of IpgB(1) -induced Yersinia invasion and (iii) failure of YopE-mediated suppression of IpgB(1) -induced Yersinia invasion, presumably due to preferential inhibition of RhoG by YopE GAP function. By infecting polarized MDCK cells we could demonstrate that Map or IpgB(1) but not IpgB(2) affects cell monolayer integrity. In summary, the Yersinia toolbox is suitable to study cellular effects of effector proteins of diverse bacterial species separately or in combination in the context of bacterial T3SS-mediated injection.
Collapse
Affiliation(s)
- Stefan Wölke
- Max von Pettenkofer Institut, LMU Munich, Pettenkofer Straße 9A, 80336 Munich, Germany
| | | | | |
Collapse
|
14
|
Terry S, Nie M, Matter K, Balda MS. Rho signaling and tight junction functions. Physiology (Bethesda) 2010; 25:16-26. [PMID: 20134025 DOI: 10.1152/physiol.00034.2009] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tight junctions are heteromeric protein complexes that act as signaling centers by mediating the bidirectional transmission of information between the environment and the cell interior to control paracellular permeability and differentiation. Insight into tight junction-associated signaling mechanisms is of fundamental importance for our understanding of the physiology of epithelia and endothelia in health and disease.
Collapse
Affiliation(s)
- Steve Terry
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | | | | | |
Collapse
|
15
|
Young EF, Quinn DA, Davies RJ. Interrogation of the rat mammary gland using intraductal impedance spectroscopy. Physiol Meas 2010; 31:451-60. [PMID: 20177141 DOI: 10.1088/0967-3334/31/3/012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Extant technologies for the detection of breast cancer exploit changes in the morphology of the mammary ductal epithelial network and can involve ionizing radiation. Intraductal surveillance of mammary epithelium has the potential to allow for earlier detection based on changes in function of the epithelium. This study investigated the feasibility of using intraductal impedance spectroscopy (IIS) to assess changes in resistance in the mammary epithelium in a small group of female rats in resting, pregnant and ultimately lactating states. In resting rats, intraductal surveillance was able to detect only a single resistive capacitance (RC). In pregnant animals, a second RC became evident in the frequency range between 1 and 190 Hz. The real resistance of this low frequency RC increased when measurements were made after the animals had begun lactating. Equivalent circuit modeling revealed this increase to be a 1.7-fold change from pregnancy to lactation. A model of tight junction closure in the context of ductal expansion is proposed. These results suggest that physiologic measurements can be made in rodent mammary epithelium using this technique allowing for assessment of function in normal and disease states.
Collapse
Affiliation(s)
- E F Young
- Department of Surgery, Hackensack University Medical Center, Hackensack, NJ, USA.
| | | | | |
Collapse
|
16
|
LeSimple P, Liao J, Robert R, Gruenert DC, Hanrahan JW. Cystic fibrosis transmembrane conductance regulator trafficking modulates the barrier function of airway epithelial cell monolayers. J Physiol 2010; 588:1195-209. [PMID: 20156845 DOI: 10.1113/jphysiol.2009.182246] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is an integral membrane glycoprotein which functions as an anion channel and influences diverse cellular processes. We studied its role in the development of epithelial tightness by expressing wild-type (WT-CFTR) or mutant (Delta F508-CFTR) CFTR in human airway epithelial cell monolayers cultured at the air-liquid interface. Green fluorescent protein (GFP)-tagged WT or Delta F508 constructs were expressed in the CF bronchial cell line CFBE41o(-) using adenoviruses, and the results were compared with those obtained using CFBE41o(-) lines stably complemented with wild-type or mutant CFTR. As predicted, GFP-Delta WT-CFTR reached the apical membrane whereas GFP-F508-CFTR was only detected intracellularly. Although CFTR expression would be expected to reduce transepithelial resistance (TER), expressing GFP-CFTR significantly increased the TER of CFBE41o(-) monolayers whilst GFP-Delta F508-CFTR had no effect. Similar results were obtained with cell lines stably overexpressing Delta F508-CFTR or WT-CFTR. Preincubating Delta F508-CFTR monolayers at 29 degrees C reduced mannitol permeability and restored TER, and the effect on TER was reversible during temperature oscillations. Expression of GFP-Delta F508-CFTR or GFP-WT-CFTR in a cell line already containing endogenous WT-CFTR (Calu-3) did not alter TER. The CFTR- and temperature-dependence of TER were not affected by the CFTR inhibitor CFTR(inh)172 or low-chloride medium; therefore the effect of CFTR on barrier function was unrelated to its ion channel activity. Modulation of TER was blunted but not eliminated by genistein, implying the involvement of tyrosine phosphorylation and other mechanisms. Modulation of CFTR trafficking was correlated with an increase in tight junction depth. The results suggest that CFTR trafficking is required for the normal organisation and function of tight junctions. A reduction in barrier function caused by endoplasmic reticulum retention of Delta F508-CFTR may contribute to fluid hyperabsorption in CF airways.
Collapse
Affiliation(s)
- Pierre LeSimple
- Department of Physiology, McGill University, Montréal, QC, Canada
| | | | | | | | | |
Collapse
|
17
|
Steed E, Balda MS, Matter K. Dynamics and functions of tight junctions. Trends Cell Biol 2010; 20:142-9. [PMID: 20061152 DOI: 10.1016/j.tcb.2009.12.002] [Citation(s) in RCA: 299] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 12/02/2009] [Accepted: 12/02/2009] [Indexed: 12/13/2022]
Abstract
Tight junctions are intercellular adhesion complexes in vertebrates that are required for the formation of functional epithelial and endothelial barriers. Their morphological appearance and biochemical composition, that includes large multimeric protein complexes, have long fostered the belief that they are relatively rigid, non-dynamic structures. Recent observations now suggest that at least some junctional elements and proteins can be very dynamic, and that such dynamic properties are important for different tight junction functions ranging from the regulation of paracellular permeability to junction-associated signalling mechanisms that guide cell behaviour. Combining such dynamic properties with existing tight junction models will help us to advance our understanding of the molecular mechanisms that underlie the functional properties of tight junctions.
Collapse
Affiliation(s)
- Emily Steed
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK
| | | | | |
Collapse
|
18
|
Swystun VA, Renaux B, Moreau F, Wen S, Peplowski MA, Hollenberg MD, MacNaughton WK. Serine proteases decrease intestinal epithelial ion permeability by activation of protein kinase Czeta. Am J Physiol Gastrointest Liver Physiol 2009; 297:G60-70. [PMID: 19460843 DOI: 10.1152/ajpgi.00096.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Epithelial permeability to ions and larger molecules in the gut is essential for fluid balance, and its dysregulation contributes to intestinal pathology. We investigated the effect of digestive serine proteases on epithelial paracellular permeability. Trypsin, chymotrypsin, and elastase elicited sustained increases in transepithelial resistance (R(TE)) in polarized monolayers of three intestinal epithelial cell lines. This effect was reflected by decreases in paracellular conductances of Na+ and Cl- and a concomitant decrease in permeability to 3,000 molecular weight dextran. The enzyme activities of the proteases were required, yet activators of known protease-activated receptors (PARs) did not reproduce the effect of these proteases on R(TE). PKCzeta isoform-specific inhibitor significantly reduced the trypsin-induced increase in R(TE) whereas PKCzeta activity was increased in cells treated with trypsin and chymotrypsin compared with control cells; this activity was reduced to control levels in the presence of PKCzeta-specific inhibitor. Ca2+ chelators and pharmacological inhibitors of cell signaling support the role for PKCzeta in the protease-induced effect. Finally, we showed that treatment with the serine proteases increased occludin immunostaining and zonula occludin-1 coimmunoprecipitation with occludin in the detergent-insoluble fraction of cell lysates, and these increases were ablated by pretreatment with PKCzeta-specific inhibitor. This finding indicates increased insertion of occludin into the cell junctional complex. These data demonstrate a role for serine proteases in the facilitation of epithelial barrier function through a mechanism that is independent of PARs and is mediated by activation of PKCzeta.
Collapse
Affiliation(s)
- Veronica A Swystun
- Inflammation Research Network, Department of Physiology and Pharmacology, University of Calgary, Calgary T2N 4N1, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Popoff MR, Geny B. Multifaceted role of Rho, Rac, Cdc42 and Ras in intercellular junctions, lessons from toxins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:797-812. [PMID: 19366594 DOI: 10.1016/j.bbamem.2009.01.011] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 01/20/2009] [Accepted: 01/22/2009] [Indexed: 12/20/2022]
Abstract
Tight junctions (TJs) and adherens junctions (AJs) are dynamic structures linked to the actin cytoskeleton, which control the paracellular permeability of epithelial and endothelial barriers. TJs and AJs are strictly regulated in a spatio-temporal manner by a complex signaling network, including Rho/Ras-GTPases, which have a pivotal role. Rho preferentially regulates TJs by controlling the contraction of apical acto-myosin filaments, whereas Rac/Cdc42 mainly coordinate the assembly-disassembly of AJ components. However, a subtle balance of Rho/Ras-GTPase activity and interplay between these molecules is required to maintain an optimal organization and function of TJs and AJs. Conversely, integrity of intercellular junctions generates signals through Rho-GTPases, which are involved in the regulation of multiple cellular processes. Rho/Ras-GTPases and the control of intercellular junctions are the target of various bacterial toxins responsible for severe diseases in man and animals, and are part of their mechanism of action. This review focuses on the regulation of TJs and AJs by Rho/Ras-GTPases through molecular approaches and bacterial toxins.
Collapse
Affiliation(s)
- Michel R Popoff
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, 75724 Paris cedex151, France.
| | | |
Collapse
|
20
|
Lee DBN, Jamgotchian N, Allen SG, Abeles MB, Ward HJ. A lipid-protein hybrid model for tight junction. Am J Physiol Renal Physiol 2008; 295:F1601-12. [PMID: 18701633 PMCID: PMC2604825 DOI: 10.1152/ajprenal.00097.2008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 08/11/2008] [Indexed: 11/22/2022] Open
Abstract
The epithelial tight junction (TJ) was first described ultrastructurally as a fusion of the outer lipid leaflets of the adjoining cell membrane bilayers (hemifusion). The discovery of an increasing number of integral TJ and TJ-associated proteins has eclipsed the original lipid-based model with the wide acceptance of a protein-centric model for the TJ. In this review, we stress the importance of lipids in TJ structure and function. A lipid-protein hybrid model accommodates a large body of information supporting the lipidic characteristics of the TJ, harmonizes with the accumulating evidence supporting the TJ as an assembly of lipid rafts, and focuses on an important, but relatively unexplored, field of lipid-protein interactions in the morphology, physiology, and pathophysiology of the TJ.
Collapse
Affiliation(s)
- David B N Lee
- Dept. of Medicine (111 VA Medical Center, 16111 Plummer St., North Hills, CA 91343, USA.
| | | | | | | | | |
Collapse
|
21
|
Korostylev A, Worzfeld T, Deng S, Friedel RH, Swiercz JM, Vodrazka P, Maier V, Hirschberg A, Ohoka Y, Inagaki S, Offermanns S, Kuner R. A functional role for semaphorin 4D/plexin B1 interactions in epithelial branching morphogenesis during organogenesis. Development 2008; 135:3333-43. [PMID: 18799546 DOI: 10.1242/dev.019760] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Semaphorins and their receptors, plexins, carry out important functions during development and disease. In contrast to the well-characterized plexin A family, however, very little is known about the functional relevance of B-type plexins in organogenesis, particularly outside the nervous system. Here, we demonstrate that plexin B1 and its ligand Sema4d are selectively expressed in epithelial and mesenchymal compartments during key steps in the genesis of some organs. This selective expression suggests a role in epithelial-mesenchymal interactions. Importantly, using the developing metanephros as a model system, we have observed that endogenously expressed and exogenously supplemented Sema4d inhibits branching morphogenesis during early stages of development of the ureteric collecting duct system. Our results further suggest that the RhoA-ROCK pathway, which is activated downstream of plexin B1, mediates these inhibitory morphogenetic effects of Sema4d and suppresses branch-promoting signalling effectors of the plexin B1 signalling complex. Finally, mice that lack plexin B1 show early anomalies in kidney development in vivo. These results identify a novel function for plexin B1 as a negative regulator of branching morphogenesis during kidney development, and suggest that the Sema4d-plexin B1 ligand-receptor pair contributes to epithelial-mesenchymal interactions during organogenesis via modulation of RhoA signalling.
Collapse
Affiliation(s)
- Alexander Korostylev
- Pharmacology Institute, Im Neuenheimer Feld 366, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Van Itallie CM, Holmes J, Bridges A, Gookin JL, Coccaro MR, Proctor W, Colegio OR, Anderson JM. The density of small tight junction pores varies among cell types and is increased by expression of claudin-2. J Cell Sci 2008; 121:298-305. [PMID: 18198187 DOI: 10.1242/jcs.021485] [Citation(s) in RCA: 293] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epithelial tight junctions contain size- and charge-selective pores that control the paracellular movement of charged and noncharged solutes. Claudins influence the charge selectivity and electrical resistance of junctions, but there is no direct evidence describing pore composition or whether pore size or density differs among cell types. To characterize paracellular pores independent of influences from charge selectivity, we profiled the ;apparent permeabilities' (P(app)) of a continuous series of noncharged polyethylene glycols (PEGs) across monolayers of five different epithelial cell lines and porcine ileum. We also characterized P(app) of high and low electrical resistance MDCK cell monolayers expressing heterologous claudins. P(app) profiling confirms that the paracellular barrier to noncharged solutes can be modeled as two distinct pathways: high-capacity small pores and a size-independent pathway allowing flux of larger solutes. All cell lines and ileum share a pore aperture of radius 4 A. Using P(app) of a PEG of radius 3.5 A to report the relative pore number provides the novel insight that pore density along the junction varies among cell types and is not necessarily related to electrical resistance. Expression of claudin-2 results in a selective increase in pore number but not size and has no effect on the permeability of PEGs that are larger than the pores; however, neither knockdown of claudin-2 nor overexpression of several other claudins altered either the number of small pores or their size. We speculate that permeability of all small solutes is proportional to pore number but that small electrolytes are subject to further selectivity by the profile of claudins expressed, explaining the dissociation between the P(app) for noncharged solutes and electrical resistance. Although claudins are likely to be components of the small pores, other factors might regulate pore number.
Collapse
|
23
|
Katsube T, Tsuji H, Onoda M. Nitric oxide attenuates hydrogen peroxide-induced barrier disruption and protein tyrosine phosphorylation in monolayers of intestinal epithelial cell. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:794-803. [PMID: 17451824 DOI: 10.1016/j.bbamcr.2007.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 03/07/2007] [Accepted: 03/09/2007] [Indexed: 12/21/2022]
Abstract
The intestinal epithelium provides a barrier to the transport of harmful luminal molecules into the systemic circulation. A dysfunctional epithelial barrier is closely associated with the pathogenesis of a variety of intestinal and systemic disorders. We investigated here the effects of nitric oxide (NO) and hydrogen peroxide (H(2)O(2)) on the barrier function of a human intestinal epithelial cell line, Caco-2. When treated with H(2)O(2), Caco-2 cell monolayers grown on permeable supports exhibited several remarkable features of barrier dysfunction as follows: a decrease in transepithelial electrical resistance, an increase in paracellular permeability to dextran, and a disruption of the intercellular junctional localization of the scaffolding protein ZO-1. In addition, an induction of tyrosine phosphorylation of numerous cellular proteins including ZO-1, E-cadherin, and beta-catenin, components of tight and adherens junctions, was observed. On the other hand, combined treatment of Caco-2 monolayers with H(2)O(2) and an NO donor (NOC5 or NOC12) relieved the damage to the barrier function and suppressed the protein tyrosine phosphorylation induced by H(2)O(2) alone. These results suggest that NO protects the barrier function of intestinal epithelia from oxidative stress by modulating some intracellular signaling pathways of protein tyrosine phosphorylation in epithelial cells.
Collapse
Affiliation(s)
- Takanori Katsube
- Radiation Effect Mechanisms Research Group, National Institute of Radiological Sciences, 4-9-1, Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan.
| | | | | |
Collapse
|
24
|
Rehder D, Iden S, Nasdala I, Wegener J, Brickwedde MKMZ, Vestweber D, Ebnet K. Junctional adhesion molecule-a participates in the formation of apico-basal polarity through different domains. Exp Cell Res 2006; 312:3389-403. [PMID: 16919624 DOI: 10.1016/j.yexcr.2006.07.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Revised: 07/06/2006] [Accepted: 07/14/2006] [Indexed: 12/21/2022]
Abstract
Junctional adhesion molecule (JAM)-A is an integral membrane protein at tight junctions of epithelial cells which associates with the cell polarity protein PAR-3. Here, we demonstrate that downregulation of JAM-A impairs the ability of MDCK II cells to form cysts in a three-dimensional matrix indicating the requirement of JAM-A for the development of apico-basal polarity. To define the regions of JAM-A important for this function, we have generated MDCK II cell lines stably expressing inducible JAM-A mutants. Mutants of JAM-A which were designed to mislocalize strongly impaired the development of cysts and the formation of functional tight junctions. Surprisingly, similar mutants that lacked the PDZ domain-binding motif at the C-terminus were still impaired in apico-basal polarity formation suggesting that additional regions within the cytoplasmic tail of JAM-A are important for the function of JAM-A. A JAM-A mutant lacking the first Ig-like domain necessary for homophilic binding localized to cell-cell contacts similar to wild-type JAM-A. However, despite this same localization, this mutant interfered with cell polarity and tight junction formation. Together our findings suggest an important role for JAM-A in the development of apico-basal polarity in epithelial cells and identify regions in JAM-A which are critical for this role.
Collapse
Affiliation(s)
- Daniela Rehder
- Institute of Cell Biology, ZMBE, University of Münster, D-48149 Münster, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Ogawa K, Wada H, Okada N, Harada I, Nakajima T, Pasquale EB, Tsuyama S. EphB2 and ephrin-B1 expressed in the adult kidney regulate the cytoarchitecture of medullary tubule cells through Rho family GTPases. J Cell Sci 2006; 119:559-70. [PMID: 16443753 DOI: 10.1242/jcs.02777] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Eph receptors and ephrin ligands are membrane-bound cell-cell communication molecules with well-defined functions in development, but their expression patterns and functions in many adult tissues are still largely unknown. We have detected substantial levels of the EphB2 and EphB6 receptors and the ephrin-B1 ligand in the adult mouse kidney by RT-PCR amplification. Immunolocalization experiments revealed that EphB2 is localized in the tubules of the inner and outer medulla and EphB6 is in the tubules of the outer medulla and cortex. By contrast, ephrin-B1 was detected in tubules throughout the whole nephron. Consistent with the overlapping expression of the EphB2 receptor and the ephrin-B1 ligand in the medulla, EphB2 is tyrosine-phosphorylated, and therefore activated, in the kidney. In the outer medulla, however, EphB2 signaling may be attenuated by the co-expressed kinase-inactive EphB6 receptor. Interestingly, we found that EphB signaling induces RhoA activation and Rac1 inactivation as well as cell retraction, enlargement of focal adhesions and prominent stress fibers in primary cultures of medullary tubule cells. These results suggest that EphB receptor signaling through Rho family GTPases regulates the cytoarchitecture and spatial organization of the tubule cells in the adult kidney medulla and, therefore, may affect the reabsorption ability of the kidney.
Collapse
Affiliation(s)
- Kazushige Ogawa
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka 599-8531, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Shi LZ, Zheng W. Establishment of an in vitro brain barrier epithelial transport system for pharmacological and toxicological study. Brain Res 2006; 1057:37-48. [PMID: 16126179 PMCID: PMC4151265 DOI: 10.1016/j.brainres.2005.07.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Revised: 07/14/2005] [Accepted: 07/18/2005] [Indexed: 11/29/2022]
Abstract
An immortalized Z310 murine choroidal epithelial cell line was recently established in this laboratory. The purposes of this study were (1) to investigate the presence of tight junction (TJ) proteins in Z310 cells and (2) to develop a Z310 cell-based in vitro brain barrier transport model. Real-time RT-PCR studies revealed that Z310 cells possess mRNAs encoding ZO-1, -2, and -3, claudin-1, -2, -4, and -8, occludin, and connexin-32. Confocal microscopic analyses confirmed the presence of claudin-1 and ZO-1 in Z310 cells at cell-cell contact sites. When Z310 cells were grown on a two-chamber Transwell device, the [14C]sucrose permeability coefficient and transepithelial electrical resistance (TEER) across the cell monolayer were 6 x 10(-4) cm/min and 61 omega-cm2, respectively. To improve the tightness of Z310 barrier, the cells were cultured in astrocyte-conditioned medium (ACM), or in the presence of eicosapentaenoic acids (EPA, 10 microM), epidermal growth factor (EGF, 100 ng/mL), or dexamethasone (1 microM) in the growth medium. Treatment with ACM, EPA, EGF and dexamethasone significantly increased the TEER by 33%, 38%, 40%, and 50% above controls, respectively. However, only dexamethasone significantly reduced [14C]sucrose paracellular permeability (-231% of controls). These data suggest that Z310 cells possess the TJ proteins. The presence of dexamethasone in the growth medium improves the tightness of Z310 cell monolayer to the level better than that of the primary culture of choroidal epithelial cells. The Z310 cell-based in vitro model appears to be suitable for transepithelial transport study of drugs and toxicants.
Collapse
Affiliation(s)
| | - Wei Zheng
- Corresponding author. Fax: +1 765 496 1377. (W. Zheng)
| |
Collapse
|
27
|
Aijaz S, Balda MS, Matter K. Tight junctions: molecular architecture and function. INTERNATIONAL REVIEW OF CYTOLOGY 2006; 248:261-98. [PMID: 16487793 DOI: 10.1016/s0074-7696(06)48005-0] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tight junctions are the most apical component of the epithelial junctional complex and are crucial for the formation and functioning of epithelial and endothelial barriers. They regulate selective diffusion of ions and solutes along the paracellular pathway and restrict apical/basolateral intramembrane diffusion of lipids. Research over the past years provided much insight into the molecular composition of tight junctions, and we are starting to understand the mechanisms that permit selective paracellular diffusion. Moreover, a complex network of proteins has been identified at tight junctions that is based on cytoskeleton-linked adaptors that recruit and thereby often regulate different types of signaling components that regulate epithelial proliferation, differentiation, and polarization.
Collapse
Affiliation(s)
- Saima Aijaz
- Division of Cell Biology, Institute of Ophthalmology, University College London, London, United Kingdom
| | | | | |
Collapse
|
28
|
Matsuzawa T, Kuwae A, Abe A. Enteropathogenic Escherichia coli type III effectors EspG and EspG2 alter epithelial paracellular permeability. Infect Immun 2005; 73:6283-9. [PMID: 16177299 PMCID: PMC1230889 DOI: 10.1128/iai.73.10.6283-6289.2005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) delivers a subset of effectors into host cells via a type III secretion system. Here we show that the type III effector EspG and its homologue EspG2 alter epithelial paracellular permeability. When MDCK cells were infected with wild-type (WT) EPEC, RhoA was activated, and this event was dependent on the delivery of either EspG or EspG2 into host cells. In contrast, a loss of transepithelial electrical resistance and ZO-1 disruption were induced by infection with an espG/espG2 double-knockout mutant, as was the case with the WT EPEC, indicating that EspG/EspG2 is not involved in the disruption of tight junctions during EPEC infection. Although EspG- and EspG2-expressing MDCK cells exhibited normal overall morphology and maintained fully assembled tight junctions, the paracellular permeability to 4-kDa dextran, but not the paracellular permeability to 500-kDa dextran, was greatly increased. This report reveals for the first time that a pathogen can regulate the size-selective paracellular permeability of epithelial cells in order to elicit a disease process.
Collapse
Affiliation(s)
- Takeshi Matsuzawa
- Laboratory of Bacterial Infection, Kitasato Institute for Life Sciences, Kitasato University, Minato-ku, Tokyo, Japan
| | | | | |
Collapse
|
29
|
Hawkins BT, Davis TP. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev 2005; 57:173-85. [PMID: 15914466 DOI: 10.1124/pr.57.2.4] [Citation(s) in RCA: 1824] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The blood-brain barrier (BBB) is the regulated interface between the peripheral circulation and the central nervous system (CNS). Although originally observed by Paul Ehrlich in 1885, the nature of the BBB was debated well into the 20th century. The anatomical substrate of the BBB is the cerebral microvascular endothelium, which, together with astrocytes, pericytes, neurons, and the extracellular matrix, constitute a "neurovascular unit" that is essential for the health and function of the CNS. Tight junctions (TJ) between endothelial cells of the BBB restrict paracellular diffusion of water-soluble substances from blood to brain. The TJ is an intricate complex of transmembrane (junctional adhesion molecule-1, occludin, and claudins) and cytoplasmic (zonula occludens-1 and -2, cingulin, AF-6, and 7H6) proteins linked to the actin cytoskeleton. The expression and subcellular localization of TJ proteins are modulated by several intrinsic signaling pathways, including those involving calcium, phosphorylation, and G-proteins. Disruption of BBB TJ by disease or drugs can lead to impaired BBB function and thus compromise the CNS. Therefore, understanding how BBB TJ might be affected by various factors holds significant promise for the prevention and treatment of neurological diseases.
Collapse
Affiliation(s)
- Brian T Hawkins
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | | |
Collapse
|
30
|
Gon Y, Wood MR, Kiosses WB, Jo E, Sanna MG, Chun J, Rosen H. S1P3 receptor-induced reorganization of epithelial tight junctions compromises lung barrier integrity and is potentiated by TNF. Proc Natl Acad Sci U S A 2005; 102:9270-5. [PMID: 15968000 PMCID: PMC1166603 DOI: 10.1073/pnas.0501997102] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pulmonary pathologies including adult respiratory distress syndrome are characterized by disruption of pulmonary integrity and edema compromising respiratory function. Sphingosine 1-phosphate (S1P) is a lipid mediator synthesized and/or stored in mast cells, platelets, and epithelial cells, with production up-regulated by the proinflammatory cytokines IL-1 and TNF. S1P administration via the airways but not via the vasculature induces lung leakage. Using receptor-null mice, we show that S1P, acting on S1P3 receptor expressed on both type I and type II alveolar epithelial cells but not vascular endothelium, induces pulmonary edema by acute tight junction opening. WT but not S1P3-null mice showed disruption of pulmonary epithelial tight junctions and the appearance of paracellular gaps between epithelial cells by electron microscopy within 1 h of airways exposure to S1P. We further show by fluorescence microscopy that S1P induced rapid loss of ZO-1 reactivity, an essential component of the cytoplasmic plaque associated with tight junctions, as well as of the tetraspannin Claudin-18, an integral membrane organizer of tight junctions. S1P shows synergistic activity with the proinflammatory cytokine TNF, showing both pulmonary edema and mortality at subthreshold S1P doses. Specifically, preexposure of mice to subthreshold doses of TNF, which alone induced no lung edema, exacerbated S1P-induced edema and impaired survival. S1P, acting through S1P3, regulates epithelial integrity and acts additively with TNF in compromising respiratory barrier function. Because S1P3-null mice are resistant to S1P-induced pulmonary leakage, either alone or in the presence of TNF, S1P3 antagonism may be useful in protecting epithelial integrity in pulmonary disease.
Collapse
MESH Headings
- Animals
- Cell Membrane/metabolism
- Cell Nucleus/metabolism
- Chromatography, Liquid
- Claudins
- Cytoplasm/metabolism
- Endothelium, Vascular/metabolism
- Epithelial Cells/metabolism
- Epithelium/metabolism
- Guanosine Triphosphate/metabolism
- Homozygote
- Inflammation
- Interleukin-1/metabolism
- Ligands
- Lung/metabolism
- Lung Diseases/metabolism
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Electron
- Microscopy, Fluorescence
- Models, Biological
- Permeability
- Protein Binding
- RNA, Messenger/metabolism
- Receptors, Lysosphingolipid/metabolism
- Receptors, Lysosphingolipid/physiology
- Tight Junctions/metabolism
- Time Factors
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Yasuhiro Gon
- Department of Immunology, Core Microscopy Facility, Helen L. Dorris Child and Adolescent Neuro-Psychiatric Disorder Institute, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Ikari A, Hirai N, Shiroma M, Harada H, Sakai H, Hayashi H, Suzuki Y, Degawa M, Takagi K. Association of Paracellin-1 with ZO-1 Augments the Reabsorption of Divalent Cations in Renal Epithelial Cells. J Biol Chem 2004; 279:54826-32. [PMID: 15496416 DOI: 10.1074/jbc.m406331200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Paracellin-1 (PCLN-1) belongs to the claudin family of tight junction proteins and possibly plays a critical role in the reabsorption of magnesium and calcium. So far, the physiological properties of PCLN-1 have not been clarified. In the present study, we investigated whether PCLN-1 is associated with ZO-1. We also investigated whether (45)Ca(2+) transport across the paracellular barrier is affected by this association. In vitro binding analysis using glutathione S-transferase fusion protein showed that the C-terminal TRV sequence, especially Thr and Val residues, of PCLN-1 interacts with ZO-1. Next, PCLN-1 was stably expressed in Madin-Darby canine kidney cells using a FLAG tagging vector. ZO-1 was co-immunoprecipitated with the wild-type PCLN-1 and the alanine substitution (TAV) mutant. However, mutants of the deletion (Delta TRV) and the alanine substitution (ARV and TRA) inhibited the association of PCLN-1 with ZO-1. Confocal immunofluorescence demonstrated that the wild-type PCLN-1 and the TAV mutant localized in the tight junction along with ZO-1, but the Delta TRV, ARV, and TRA mutants were widely distributed in the lateral membrane including the tight junction area. Interestingly, monolayers of cells expressing the wild-type PCLN-1 and the TAV mutant showed higher activities of (45)Ca(2+) transport from apical to basal compartments, compared with those expressing the Delta TRV, ARV, and TRA mutants and the mock cells. (45)Ca(2+) transport was inhibited by increased magnesium concentration suggesting that magnesium and calcium were competitively transported by PCLN-1. It was noted that a positive electrical potential gradient enhanced (45)Ca(2+) transport from apical to basal compartments without affecting the opposite direction of transport. Thus, PCLN-1 localizes to the tight junction followed by association with ZO-1, and the PCLN-1.ZO-1 complex may play an essential role in the reabsorption of divalent cations in renal epithelial cells.
Collapse
Affiliation(s)
- Akira Ikari
- Department of Environmental Biochemistry and Toxicology, University of Shizuoka, 52-1 Yada, Shizuoka 422-8526, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Harhaj NS, Antonetti DA. Regulation of tight junctions and loss of barrier function in pathophysiology. Int J Biochem Cell Biol 2004; 36:1206-37. [PMID: 15109567 DOI: 10.1016/j.biocel.2003.08.007] [Citation(s) in RCA: 404] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2003] [Accepted: 08/21/2003] [Indexed: 12/16/2022]
Abstract
The mechanism by which epithelial and endothelial cells interact to form polarized tissue is of fundamental importance to multicellular organisms. Dysregulation of these barriers occurs in a variety of diseases, destroying the normal cellular environments and leading to organ failure. Increased levels of growth factors are a common characteristic of diseases exhibiting tissue permeability, suggesting that growth factors play a direct role in elevating permeability. Of particular concern for this laboratory, increased expression of vascular endothelial growth factor may enhance vascular permeability in diabetic retinopathy, leading to vision impairment and blindness. However, the mechanism by which growth factors increase permeability is unclear. Polarized cells form strong barriers through the development of tight junctions, which are specialized regions of the junctional complex. Tight junctions are composed of three types of transmembrane proteins, a number of peripheral membrane structural proteins, and are associated with a variety of regulatory proteins. Recent data suggest that growth factor-stimulated alterations in tight junctions contribute to permeability in a variety of disease states. The goal of this review was to elucidate potential mechanisms by which elevated growth factors elicit deregulated paracellular permeability via altered regulation of tight junctions, with particular emphasis on the tight junction proteins occludin and ZO-1, protein kinase C signaling, and endocytosis of junctional proteins. Understanding the molecular mechanisms underlying growth factor-mediated regulation of tight junctions will facilitate the development of novel treatments for diseases such as brain tumors, diabetic retinopathy and other diseases with compromised tight junction barriers.
Collapse
Affiliation(s)
- Nicole S Harhaj
- Penn State Retina Research Group, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | |
Collapse
|
33
|
Abstract
Multicellular organisms are separated from the external environment by a layer of epithelial cells whose integrity is maintained by intercellular junctional complexes composed of tight junctions, adherens junctions, and desmosomes, whereas gap junctions provide for intercellular communication. The aim of this review is to present an updated overview of recent developments in the area of tight junction biology. In a relatively short time, our knowledge of the tight junction has evolved from a relatively simple view of it being a permeability barrier in the paracellular space and a fence in the plane of the plasma membrane to one of it acting as a multicomponent, multifunctional complex that is involved in regulating numerous and diverse cell functions. A group of integral membrane proteins-occludin, claudins, and junction adhesion molecules-interact with an increasingly complex array of tight junction plaque proteins not only to regulate paracellular solute and water flux but also to integrate such diverse processes as gene transcription, tumor suppression, cell proliferation, and cell polarity.
Collapse
Affiliation(s)
- Eveline E Schneeberger
- Molecular Pathology Unit, Massachusetts General Hospital East, Charlestown, MA 02129, USA.
| | | |
Collapse
|
34
|
Abstract
Epithelial and endothelial cells are joined to each other via a set of intercellular junctions that differ in their morphological appearance, composition, and function. The tight junction or zonula occludens is the intercellular junction that regulates diffusion between cells and therefore allows endothelia and epithelia to form cellular barriers that separate compartments of different composition. This intercellular gate formed by tight junctions is not only highly regulated but is size- and ion-selective and, hence, represents a semipermeable diffusion barrier. In epithelia, tight junctions form a morphological and functional border between the apical and basolateral cell surface domains. They directly contribute to the maintenance of cell surface polarity by forming a fence that prevents apical/basolateral diffusion of lipids in the outer leaflet of the plasma membrane. Here we describe a set of assays that allow the analysis of tight junctions to determine their integrity and functional state.
Collapse
Affiliation(s)
- Karl Matter
- Division of Cell Biology, Institute of Ophthalmology, University College London, Bath Street, EC1V 9EL, London, UK.
| | | |
Collapse
|
35
|
Rubenstein NM, Guan Y, Woo PL, Firestone GL. Glucocorticoid down-regulation of RhoA is required for the steroid-induced organization of the junctional complex and tight junction formation in rat mammary epithelial tumor cells. J Biol Chem 2003; 278:10353-60. [PMID: 12525486 DOI: 10.1074/jbc.m213121200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In Con8 mammary epithelial tumor cells, we have documented previously that the synthetic glucocorticoid dexamethasone induces the reorganization of the tight junction and adherens junction (apical junction) and stimulates the monolayer transepithelial electrical resistance (TER), which is a reliable in vitro measurement of tight junction sealing. Western blots demonstrated that dexamethasone treatment down-regulated the level of the RhoA small GTPase prior to the stimulation of the monolayer TER. To test the role of RhoA in the steroid regulation of apical junction dynamics functionally, RhoA levels were altered in Con8 cells by transfection of either constitutively active (RhoA.V14) or dominant negative (RhoA.DN19) forms of RhoA. Ectopic expression of constitutively active RhoA disrupted the dexamethasone-stimulated localization of zonula occludens-1 and beta-catenin to sites of cell-cell contact, inhibited tight junction sealing, and prevented the complete formation of the F-actin ring structure at the apical side of the cell monolayer. In a complementary manner, dominant negative RhoA caused a precocious organization of the tight junction, adherens junction, and the F-actin rings in the absence of steroid, whereas the monolayer TER remained glucocorticoid-responsive. Taken together, our results demonstrate that the glucocorticoid down-regulation of RhoA is a required step in the steroid signaling pathway which controls the organization of the apical junctional complex and the actin cytoskeleton in mammary epithelial cells.
Collapse
Affiliation(s)
- Nicola M Rubenstein
- Department of Molecular and Cell Biology and the Cancer Research Laboratory, University of California at Berkeley, 94720-3200, USA
| | | | | | | |
Collapse
|
36
|
Benais-Pont G, Punn A, Flores-Maldonado C, Eckert J, Raposo G, Fleming TP, Cereijido M, Balda MS, Matter K. Identification of a tight junction-associated guanine nucleotide exchange factor that activates Rho and regulates paracellular permeability. J Cell Biol 2003; 160:729-40. [PMID: 12604587 PMCID: PMC2173357 DOI: 10.1083/jcb.200211047] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Rho family GTPases are important regulators of epithelial tight junctions (TJs); however, little is known about how the GTPases themselves are controlled during TJ assembly and function. We have identified and cloned a canine guanine nucleotide exchange factor (GEF) of the Dbl family of proto-oncogenes that activates Rho and associates with TJs. Based on sequence similarity searches and immunological and functional data, this protein is the canine homologue of human GEF-H1 and mouse Lfc, two previously identified Rho-specific exchange factors known to associate with microtubules in nonpolarized cells. In agreement with these observations, immunofluorescence of proliferating MDCK cells revealed that the endogenous canine GEF-H1/Lfc associates with mitotic spindles. Functional analysis based on overexpression and RNA interference in polarized MDCK cells revealed that this exchange factor for Rho regulates paracellular permeability of small hydrophilic tracers. Although overexpression resulted in increased size-selective paracellular permeability, such cell lines exhibited a normal overall morphology and formed fully assembled TJs as determined by measuring transepithelial resistance and by immunofluorescence and freeze-fracture analysis. These data indicate that GEF-H1/Lfc is a component of TJs and functions in the regulation of epithelial permeability.
Collapse
Affiliation(s)
- Gaelle Benais-Pont
- Department of Cell Biology, University of Geneva, 1211 Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Tight junctions have long been regarded as simple barriers that separate compartments of different compositions, but recent research indicates that different types of signalling proteins and transduction pathways are associated with these junctions. They receive and convert signals from the cell interior to regulate junction assembly and function, and transmit signals to the cell interior to modulate gene expression and cell behaviour.
Collapse
Affiliation(s)
- Karl Matter
- Division of Cell Biology, Institute of Ophthalmology, University College London, London, UK
| | | |
Collapse
|
38
|
Abstract
Rho GTPases, such as Rho, Rac and Cdc42, are known to regulate many cellular processes including cell movement and cell adhesion. While the cellular events of germ cell movement are crucial to spermatogenesis since developing germ cells must migrate progressively from the basal to the adluminal compartment but remain attached to the seminiferous epithelium, the physiological significance of Rho GTPases in spermatogenesis remains largely unexplored. This paper reviews some recent findings on Rho GTPases in the field with emphasis on the studies in the testis, upon which future studies can be designed to delineate the role of Rho GTPases in spermatogenesis.
Collapse
Affiliation(s)
- Wing-Yee Lui
- Population Council, Center for Biomedical Research, 1230 York Avenue, New York, NY 10021, USA
| | | | | |
Collapse
|
39
|
Hopkins AM, Walsh SV, Verkade P, Boquet P, Nusrat A. Constitutive activation of Rho proteins by CNF-1 influences tight junction structure and epithelial barrier function. J Cell Sci 2003; 116:725-42. [PMID: 12538773 DOI: 10.1242/jcs.00300] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The apical-most epithelial intercellular junction, referred to as the tight junction (TJ), regulates paracellular solute flux in diverse physiological and pathological states. TJ affiliations with the apical filamentous actin (F-actin) cytoskeleton are crucial in regulating TJ function. F-actin organization is influenced by the Rho GTPase family, which also controls TJ function. To explore the role of Rho GTPases in regulating TJ structure and function, we utilized Escherichia coli cytotoxic necrotizing factor-1 (CNF-1) as a tool to activate constitutively Rho, Rac and Cdc42 signaling in T84 polarized intestinal epithelial monolayers. The biological effects of the toxin were polarized to the basolateral membrane, and included profound reductions in TJ gate function, accompanied by displacement of the TJ proteins occludin and zonula occludens-1 (ZO-1), and reorganization of junction adhesion molecule-1 (JAM-1) away from the TJ membrane. Immunogold electron microscopy revealed occludin and caveolin-1 internalization in endosomal/caveolar-like structures in CNF-treated cells. Immunofluorescence/confocal microscopy suggested that a pool of internalized occludin went to caveolae, early endosomes and recycling endosomes, but not to late endosomes. This provides a novel mechanism potentially allowing occludin to evade a degradative pathway, perhaps allowing efficient recycling back to the TJ membrane. In contrast to the TJ, the characteristic ring structure of proteins in adherens junctions (AJs) was largely preserved despite CNF-1 treatment. CNF-1 also induced displacement of a TJ-associated pool of phosphorylated myosin light chain (p-MLC), which is normally also linked to the F-actin contractile machinery in epithelial cells. The apical perjunctional F-actin ring itself was maintained even after toxin exposure, but there was a striking effacement of microvillous F-actin and its binding protein, villin, from the same plane. However, basal F-actin stress fibers became prominent and cabled following basolateral CNF-1 treatment, and the focal adhesion protein paxillin was tyrosine phosphorylated. This indicates differences in Rho GTPase-mediated control of distinct F-actin pools in polarized cells. Functionally, CNF-1 profoundly impaired TJ/AJ assembly in calcium switch assays. Re-localization of occludin but not E-cadherin along the lateral membrane during junctional reassembly was severely impaired by the toxin. A balance between activity and quiescence of Rho GTPases appears crucial for both the generation and maintenance of optimal epithelial barrier function. Overactivation of Rho, Rac and Cdc42 with CNF-1 seems to mirror key barrier-function disruptions previously reported for inactivation of RhoA.
Collapse
Affiliation(s)
- Ann M Hopkins
- Department of Pathology and Laboratory Medicine, Emory University, Whitehead Biomedical Research Building, Atlanta, GA 30322, USA.
| | | | | | | | | |
Collapse
|
40
|
Harhaj NS, Barber AJ, Antonetti DA. Platelet-derived growth factor mediates tight junction redistribution and increases permeability in MDCK cells. J Cell Physiol 2002; 193:349-64. [PMID: 12384987 DOI: 10.1002/jcp.10183] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Increased tissue permeability is a common characteristic of a number of diseases such as pulmonary edema, inflammatory bowel disease, several kidney diseases, diabetic retinopathy, and tumors. We hypothesized that growth factors increase permeability by redistribution of tight junction proteins away from the cell border. To investigate mechanisms of growth factor-mediated permeability, we examined the effect of platelet derived growth factor (PDGF) on Madin-Darby canine kidney (MDCK) cell tight junction protein distribution and on permeability. PDGF altered the cellular distribution of occludin and ZO-1 from the cell border to the cytoplasm and increased permeability to 70 kDa dextran in a concentration-dependent manner. Treatment of MDCK cells with PDGF prior to fixation allowed binding of the lectin concanavalin A to the basement membrane of fixed cells, while binding was prevented in untreated control monolayers, implying that PDGF induced the formation of a paracellular transport pathway. Cell fractionation experiments with PDGF-treated cells revealed a novel occludin-containing low-density, detergent resistant subcellular structure, which increased in the buoyant fractions relative to occludin in the pellet in a time- and concentration-dependent manner. Immunocytochemistry revealed that a pool of internalized occludin co-labels with the early endosome marker, EEA1, suggesting that PDGF may stimulate occludin to enter an endosomal pathway. PDGF may act as a permeabilizing agent by moving tight junction proteins away from the cell border in discrete microdomains, and the effects of PDGF on permeability and tight junction protein distribution may model the regulation of epithelial and endothelial barrier properties by other peptide growth factors.
Collapse
Affiliation(s)
- Nicole S Harhaj
- The Penn State Retina Research Group, Penn State College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | |
Collapse
|
41
|
Gopalakrishnan S, Dunn KW, Marrs JA. Rac1, but not RhoA, signaling protects epithelial adherens junction assembly during ATP depletion. Am J Physiol Cell Physiol 2002; 283:C261-72. [PMID: 12055095 DOI: 10.1152/ajpcell.00604.2001] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rho family GTPase signaling regulates actin cytoskeleton and junctional complex assembly. Our previous work showed that RhoA signaling protects tight junctions from damage during ATP depletion. Here, we examined whether RhoA GTPase signaling protects adherens junction assembly during ATP depletion. Despite specific RhoA signaling- and ATP depletion-induced effects on adherens junction assembly, RhoA signaling did not alter adherens junction disassembly rates during ATP depletion. This shows that RhoA signaling specifically protects tight junctions from damage during ATP depletion. Rac1 GTPase signaling also regulates adherens junction assembly and therefore may regulate adherens junction assembly during ATP depletion. Indeed, we found that Rac1 signaling protects adherens junctions from damage during ATP depletion. Adherens junctions are regulated by various GTPases, including RhoA and Rac1, but adherens junctions are specifically protected by Rac1 signaling.
Collapse
Affiliation(s)
- Shobha Gopalakrishnan
- Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana 46202-5116, USA
| | | | | |
Collapse
|
42
|
Ramsey CR, Berndt TJ, Knox FG. Indomethacin blocks enhanced paracellular backflux in proximal tubules. J Am Soc Nephrol 2002; 13:1449-54. [PMID: 12039973 DOI: 10.1097/01.asn.0000015616.97847.0c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Renal interstitial hydrostatic pressure (RIHP) is a link between increased arterial BP and natriuresis. The mechanism whereby increases in RIHP inhibits sodium and water transport across the mammalian proximal tubule epithelium may involve changes in flux across the tight junction of the proximal tubule. The purpose of this study was to determine the effects of increases in RIHP and inhibition of cyclooxygenase on paracellular backflux of an extracellular marker from the renal interstitium into the proximal tubule of the rat. During in vivo microperfusion of proximal tubules, the extracellular tracer of paracellular flux, lanthanum (La), was infused directly into the renal interstitium via a chronically implanted matrix. The net paracellular interstitium-to-lumen lanthanum backflux was measured before and after direct renal interstitial volume expansion (DRIVE) in the absence and presence of indomethacin. DRIVE significantly increased RIHP by 37% (Delta1.8 +/- 0.2 mmHg) and interstitium-to-lumen La backflux by 32% (Delta40.2 +/- 16.6 pg/min per mm), and it significantly decreased proximal reabsorption by 27% (Delta-7.7 +/- 3.8 nl/min; n = 6). In indomethacin-treated rats (n = 6), DRIVE again significantly increased RIHP by 40% (Delta1.9 +/- 0.2 mmHg), but it did not increase La backflux (Delta-39.0 +/- 24.4 pg/min per mm) or significantly decrease proximal reabsorption (Delta1.2 +/- 2.3 nl/min). These results demonstrate that increased RIHP increases paracellular backflux of lanthanum from the renal interstitium to the proximal tubule lumen in association with decreases in proximal reabsorption. Furthermore, indomethacin blocks the effects of increased RIHP on proximal reabsorption and paracellular backflux of lanthanum through the intercellular tight junctions of the proximal tubule epithelium.
Collapse
Affiliation(s)
- Carla R Ramsey
- Department of Internal Medicine and Physiology and Biophysics, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
43
|
Watson CJ, Rowland M, Warhurst G. Functional modeling of tight junctions in intestinal cell monolayers using polyethylene glycol oligomers. Am J Physiol Cell Physiol 2001; 281:C388-97. [PMID: 11443038 DOI: 10.1152/ajpcell.2001.281.2.c388] [Citation(s) in RCA: 175] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite significant advances in the characterization of tight junction (TJ) proteins, little is known about how molecular changes relate to function due primarily to the limitations of conventional paracellular probes. To address this, the paracellular pathway in Caco-2 and T84 cell lines was profiled by measuring the permeabilities of 24 polyethylene glycols (PEG) of increasing molecular radius (3.5--7.4 A) analyzed by mass spectrometry. When combined with a paracellular sieving model, these data provided quantitative descriptors of the pathway under control conditions and after exposure to TJ modulators. PEG profiles in both cell lines conformed to a biphasic process involving a restrictive pore (radius 4.3--4.5 A) and a nonrestrictive component responsible for permeability of larger molecules. PEG profiling revealed significant differences between the effects of EGTA and sodium caprate (C10). The restrictive component of EGTA-treated cells lost all size discrimination due to an increase in pore radius. Sodium caprate had no effect on pore radius but increased permeability via a different mechanism possibly involving increased numbers of functional pores. PEG profiling provides a useful tool for probing the functional regulation of the paracellular route.
Collapse
Affiliation(s)
- C J Watson
- Gut Barrier Group, Section of Gastrointestinal Science, Clinical Division I, University of Manchester, Hope Hospital, Salford M6 8HD, United Kingdom
| | | | | |
Collapse
|
44
|
Van Itallie C, Rahner C, Anderson JM. Regulated expression of claudin-4 decreases paracellular conductance through a selective decrease in sodium permeability. J Clin Invest 2001; 107:1319-27. [PMID: 11375422 PMCID: PMC209303 DOI: 10.1172/jci12464] [Citation(s) in RCA: 475] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tight junctions regulate paracellular conductance and ionic selectivity. These properties vary among epithelia but the molecular basis of this variation remains unknown. To test whether members of the claudin family of tight junction proteins influence paracellular ionic selectivity, we expressed human claudin-4 in cultured MDCK cells using an inducible promoter. Overexpression increased the complexity of tight junction strands visible by freeze-fracture microscopy without affecting the levels of claudin-1, -2, or -3, occludin, or ZO-1. A decrease in conductance correlated directly with the kinetics of claudin-4 induction. Dilution potentials revealed that the decrease in paracellular conductance resulted from a selective decrease in Na(+) permeability without a significant effect on Cl(-) permeability. Flux for an uncharged solute, mannitol, and the rank order of permeabilities for the alkali metal cations were unchanged. A paracellular site for these effects was supported by the lack of apical/basal directionality of the dilution potentials, the linearity of current-voltage relationships, and the lack of influence of inhibitors of major transcellular transporters. These results provide, to our knowledge, the first direct demonstration of the ability of a claudin to influence paracellular ion selectivity and support a role for the claudins in creating selective channels through the tight-junction barrier.
Collapse
Affiliation(s)
- C Van Itallie
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8019, USA.
| | | | | |
Collapse
|
45
|
Yasui H, Katoh H, Yamaguchi Y, Aoki J, Fujita H, Mori K, Negishi M. Differential responses to nerve growth factor and epidermal growth factor in neurite outgrowth of PC12 cells are determined by Rac1 activation systems. J Biol Chem 2001; 276:15298-305. [PMID: 11278419 DOI: 10.1074/jbc.m008546200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurite outgrowth of PC12 cells is induced by nerve growth factor (NGF) but not by epidermal growth factor (EGF). This differential response has been explained by the duration of mitogen-activated protein kinase (MAPK) activation; NGF induces sustained MAPK activation but EGF leads short-lived activation. However, precise mechanisms have not yet been understood. Here we demonstrate the difference between NGF and EGF in regulation of Rac1, a small GTPase involved in neurite outgrowth, in PC12 cells. NGF phosphoinositide 3-kinase dependently induces transient activation of Rac1 and accumulation of active Rac1 at protrusion sites on the cell surface, inducing filamentous actin-rich protrusions and subsequent neurite formation in a Rac1-dependent manner. On the other hand, EGF phosphoinositide 3-kinase independently induces more transient Rac1 activation but neither accumulates active Rac1 nor forms Rac1- and filamentous actin-rich protrusions. Difference in the Rac1 localization between NGF and EGF was also observed with the localization of exogenously expressed green fluorescent protein-tagged Rac1. The Rac1-mediated protrusion by NGF is independent of MAPK cascade, but the subsequent neurite extension requires the cascade. Thus, the differential activation of Rac1 and localization of active Rac1 contribute to the difference in the ability of NGF and EGF to induce neurite outgrowth, and we propose that the MAPK cascade-independent prompt activation of Rac1 and recruitment of active Rac1 at the protrusion sites trigger the initiation of neurite formation.
Collapse
Affiliation(s)
- H Yasui
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Hollande F, Blanc EM, Bali JP, Whitehead RH, Pelegrin A, Baldwin GS, Choquet A. HGF regulates tight junctions in new nontumorigenic gastric epithelial cell line. Am J Physiol Gastrointest Liver Physiol 2001; 280:G910-21. [PMID: 11292600 DOI: 10.1152/ajpgi.2001.280.5.g910] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The regulation of intercellular adhesion by hepatocyte growth factor (HGF) was examined on a novel nontumorigenic gastric epithelial cell line (IMGE-5) derived from H-2Kb-tsA58 transgenic mice. IMGE-5 cells constitutively expressed cytokeratin 18 and HGF receptors. Under permissive conditions (33 degrees C + interferon-gamma), IMGE-5 cells proliferated rapidly but did not display membrane expression of adherens and tight junction proteins. Under nonpermissive conditions, their proliferation was decreased and they displayed a strong, localized membrane expression of E-cadherin/beta-catenin and occludin/ZO-1. HGF treatment largely prevented the targeting of ZO-1 to the tight junction and induced a significant decrease of the transepithelial resistance measured across a confluent IMGE-5 cell monolayer. HGF rapidly increased the tyrosine phosphorylation of ZO-1 and decreased its association with occludin in a phosphatidylinositol 3-kinase (PI 3-kinase)-dependent manner. PI 3-kinase was also involved in HGF-induced migration of IMGE-5 cells. Our results demonstrate that 1) HGF prevents the appearance of ZO-1 in the membrane during epithelial cell differentiation; 2) HGF causes partial relocalization of ZO-1 to the cytoplasm and nucleus and concomitantly stimulates cell dissociation and migration; and 3) IMGE-5 cells offer a useful model for the study of gastric epithelial cell differentiation.
Collapse
Affiliation(s)
- F Hollande
- Laboratoire de Biochimie des Membranes, Faculté de Pharmacie, 15 av. C. Flahault, 34060 Montpellier cedex, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Aoki J, Katoh H, Mori K, Negishi M. Rnd1, a novel rho family GTPase, induces the formation of neuritic processes in PC12 cells. Biochem Biophys Res Commun 2000; 278:604-8. [PMID: 11095956 DOI: 10.1006/bbrc.2000.3842] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Rho family GTPases have been shown to be involved in the regulation of neuronal cell morphology, including neurite extension and retraction. Rho activation leads to neurite retraction and cell rounding, whereas Rac and Cdc42 are implicated in the promotion of filopodia and lamellipodia formation in growth cones and, therefore, in neurite extension. In this study, we examined the morphological role of Rnd1, a new member of Rho family GTPases, in PC12 cells, and found that expression of Rnd1 by itself caused the formation of many neuritic processes from the cell body with disruption of the cortical actin filaments, the processes having microtubules but few filamentous actin and neurofilaments. Treatment with cytochalasin D, an inhibitor of actin polymerization, could mimic the effects of expression of Rnd1, in that this inhibitor disrupted the cortical actin filaments and induced the formation of many thin processes containing microtubules. The process formation induced by Rnd1 was inhibited by dominant negative Rac1. These results suggest that Rnd1 induces the Rac-dependent neuritic process formation in part by disruption of the cortical actin filaments.
Collapse
Affiliation(s)
- J Aoki
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | | | | | | |
Collapse
|
48
|
Katoh H, Yasui H, Yamaguchi Y, Aoki J, Fujita H, Mori K, Negishi M. Small GTPase RhoG is a key regulator for neurite outgrowth in PC12 cells. Mol Cell Biol 2000; 20:7378-87. [PMID: 10982854 PMCID: PMC86291 DOI: 10.1128/mcb.20.19.7378-7387.2000] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Rho family of small GTPases has been implicated in cytoskeletal reorganization and subsequent morphological changes in various cell types. Among them, Rac and Cdc42 have been shown to be involved in neurite outgrowth in neuronal cells. In this study, we examined the role of RhoG, another member of Rho family GTPases, in nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells. Expression of wild-type RhoG in PC12 cells induced neurite outgrowth in the absence of NGF, and the morphology of wild-type RhoG-expressing cells was similar to that of NGF-differentiated cells. Constitutively active RhoG-transfected cells extended short neurites but developed large lamellipodial or filopodial structures at the tips of neurites. RhoG-induced neurite outgrowth was inhibited by coexpression with dominant-negative Rac1 or Cdc42. In addition, expression of constitutively active RhoG elevated endogenous Rac1 and Cdc42 activities. We also found that the NGF-induced neurite outgrowth was enhanced by expression of wild-type RhoG whereas expression of dominant-negative RhoG suppressed the neurite outgrowth. Furthermore, constitutively active Ras-induced neurite outgrowth was also suppressed by dominant-negative RhoG. Taken together, these results suggest that RhoG is a key regulator in NGF-induced neurite outgrowth, acting downstream of Ras and upstream of Rac1 and Cdc42 in PC12 cells.
Collapse
Affiliation(s)
- H Katoh
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
McCarthy KM, Francis SA, McCormack JM, Lai J, Rogers RA, Skare IB, Lynch RD, Schneeberger EE. Inducible expression of claudin-1-myc but not occludin-VSV-G results in aberrant tight junction strand formation in MDCK cells. J Cell Sci 2000; 113 Pt 19:3387-98. [PMID: 10984430 DOI: 10.1242/jcs.113.19.3387] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Occludin and 18 distinct members of the claudin family are tetra-span transmembrane proteins that are localized in cell-specific tight junctions (TJs). A previous study showed that expression of chick occludin in Madin-Darby canine kidney (MDCK) cells raised transepithelial electrical resistance (TER) and, paradoxically, increased mannitol flux. In the present study, we employed epitope tagged canine occludin expression, under the control of the tetracycline repressible transactivator, to determine the extent to which the unexpected parallel increase in TER and mannitol flux was related to a structural mismatch between avian and canine occludins, which are only 50% identical. To determine whether the paradoxical changes in permeability was specific to occludin, we assessed the effect of over-expressing epitope tagged murine claudin-1. Our data revealed that over-expression of either of the epitope tagged mammalian tight junction proteins increased TER, mannitol and FITC-dextran flux. We observed a 2- and up to 5.6-fold over-expression of occludin-VSV-G and claudin-1-myc, respectively, with no change in ZO-1, endogenous occludin or claudin-1 expression. Confocal microscopy revealed that occludin-VSV-G, claudin-1-myc and ZO-1 co-localized at the TJ. In addition, claudin-1-myc formed aberrant strands along the lateral cell surface without an underlying ZO-1 scaffold. In fracture labeled replicas these strands consisted of claudin-1-myc with little accompanying occludin. These observations suggest that in epithelial cells claudin-1 can assemble into TJ strands without the participation of either ZO-1 or occludin. The proximity of the myc tag to the COOH-terminal YV sequence of claudin-1 appeared to interfere with its interaction with ZO-1, since over-expression of non-tagged claudin-1 increased TER but had a minimal effect on solute flux and no aberrant strands formed. From our data we conclude that differences in structure between avian and mammalian occludin do not account for the observed paradoxical increase in mannitol flux. Levels of ZO-1 remained unchanged despite substantial increases in induced TJ integral protein expression, suggesting that an imbalance between levels of ZO-1 and occludin or claudin-1 leads to altered regulation of pores through which non-charged solute flux occurs. We suggest that ion and solute flux are differentially regulated at the TJ.
Collapse
Affiliation(s)
- K M McCarthy
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Most human tumors are of epithelial origin, and these tumors gradually lose their epithelial character in a process termed the epithelial-mesenchymal transition. Approximately 40% of human tumors have activating mutations in one of the three RAS genes. Given these statistics, it is critically important to understand the role of Ras signaling in the epithelial-mesenchymal transition. This review considers the mechanisms and effectors through which Ras may regulate intercellular junction formation in epithelial cells. Conversely, intercellular junction proteins themselves may play a role in regulating Ras activation and signaling.
Collapse
Affiliation(s)
- J A Mercer
- McLaughlin Research Institute, 1520 23rd St South, Great Falls, MT 59405-4900, USA.
| |
Collapse
|