151
|
Hoshino M, Hashimoto S, Muramatsu T, Matsuki M, Ogiuchi H, Shimono M. Claudin rather than occludin is essential for differentiation in rat incisor odontoblasts. Oral Dis 2008; 14:606-12. [PMID: 18208478 DOI: 10.1111/j.1601-0825.2007.01427.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Many morphological and developmental studies have demonstrated the characteristics of tight junctions (TJs) between odontoblasts. However, detailed localization of TJ-associated proteins in odontoblasts and their functions has not yet been clarified. To elucidate the relationship between the establishment of TJ structures and the differentiation of odontoblasts during early dentinogenesis, we studied the expression and localization of constituent proteins of TJs (claudin-1, occludin, ZO-1 and ZO-2) between odontoblasts in rat lower incisors using Western blotting, immunofluorescence and immunoelectron microscopy. When the expression of claudin-1 increases at the distal portion of mature odontoblasts, the TJs form complex networks of strands, and odontoblasts differentiated by developing distal membrane domains and by secreting specific molecules for mineralization. We conclude that the TJs of odontoblasts may play an important role in the differentiation of odontoblasts in rat lower incisors during early dentinogenesis.
Collapse
Affiliation(s)
- M Hoshino
- Department of Oral and Maxillofacial Surgery, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
152
|
Inai T, Sengoku A, Hirose E, Iida H, Shibata Y. Claudin-7 expressed on lateral membrane of rat epididymal epithelium does not form aberrant tight junction strands. Anat Rec (Hoboken) 2008; 290:1431-8. [PMID: 17853415 DOI: 10.1002/ar.20597] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Claudins are integral membrane proteins at tight junctions (TJs) and form TJ strands. In the present study, we found that claudin-7 was localized along the entire lateral membranes of epididymal epithelium, including the apical junctional region throughout the epididymis, but claudin-8 was restricted to the apical junctional region. This finding raises the possibility that aberrant TJ strands may be formed on lateral membranes. Thus, we focused on examining whether TJ strands exist on lateral membranes of epididymal epithelium. Freeze-fracture electron microscopy showed that aberrant TJ strands were observed in only a few principal cells in all segments of the epididymis except for the initial segment, indicating that the occurrence of aberrant strands is very rare. Aberrant TJ strands were smooth and not subdivided into individual particles in the protoplasmic face, and complementary grooves in the extracellular face were almost free of particles. Aberrant TJ strands in the distal caput and corpus epididymis were accompanied by many vesicle-like structures but those in the proximal caput and cauda epididymis were not. These results suggest that most of claudin-7 in lateral membranes may exist in a nonpolymerized form and may play some different roles other than the formation of TJ strands, for example, in the formation of a pool of claudin proteins or in the reinforcement of cell adhesion.
Collapse
Affiliation(s)
- Tetsuichiro Inai
- Department of Developmental Molecular Anatomy, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
153
|
Comparative characterization of mouse rectum CMT93-I and -II cells by expression of claudin isoforms and tight junction morphology and function. Histochem Cell Biol 2007; 129:223-32. [PMID: 18034259 DOI: 10.1007/s00418-007-0360-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2007] [Indexed: 01/28/2023]
Abstract
Recent studies suggest that the morphological and physiological properties of tight junctions (TJs) are determined by the combination and mixing ratios of claudin isoforms. In this study, we tried to characterize mouse cell lines by expression of claudin isoforms to use for studying epithelial TJs by overexpression or suppression of claudin(s) in the cells and found that claudin-2 was expressed in a few mouse rectum carcinoma cells, CMT93 cells. We have isolated CMT93-I and -II cells from CMT93 cells by immunohistochemical screening for the presence or absence of claudin-2 expression. Immunofluorescence and RT-PCR analyses showed that expression of claudin-4, -6, -7 and -12 was detected in both cell lines, but claudin-2 was only expressed in CMT93-II cells. There were no differences in paracellular permeability between CMT93-I and -II cells examined by 4 kDa FITC-dextran and fluorescein sodium, or in the number of TJ strands examined by freeze-fracture electron microscopy. However, the transepithelial electrical resistance (TER) of CMT93-I cells was approximately 6.5 times higher than that of CMT93-II cells, suggesting that expression of claudin-2 may be related to decreased TER. Comparative examinations of CMT93-I and -II cells provide a clue how the combination and mixing ratios of claudin isoforms regulate the paracellular permeability.
Collapse
|
154
|
Tether and trap: regulation of membrane-raft dynamics by actin-binding proteins. Nat Rev Immunol 2007; 7:889-96. [PMID: 17948020 DOI: 10.1038/nri2193] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The existence of plasma-membrane-raft microdomains has been widely debated during the past few years. However, it is clear that during lymphocyte stimulation a lipid-based reorganization occurs at the plasma membrane, with markers of the membrane rafts being selectively recruited to key active regions of the cell. Recent reports have demonstrated that membrane-raft dynamics are controlled by proteins that are linked to the actin cytoskeleton and have suggested a new model for the plasma membrane based on protein-lipid interactions. This new and dynamic view of the plasma membrane may improve our understanding of the complex process leading to cell polarization during lymphocyte migration and activation.
Collapse
|
155
|
Sengoku A, Inai T, Shibata Y. Formation of aberrant TJ strands by overexpression of claudin-15 in MDCK II cells. Histochem Cell Biol 2007; 129:211-22. [PMID: 17989991 DOI: 10.1007/s00418-007-0354-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2007] [Indexed: 10/22/2022]
Abstract
Claudins are one of the transmembrane proteins found at tight junctions (TJs); they constitute the backbone of TJ strands and comprise a multigene family. Claudins share a YV sequence at the COOH-termini, which is considered to be a ZO-binding motif. Overexpression of claudin-15 (15CL) or claudin-15 tagged with enhanced green fluorescent protein at the NH2-terminus (EGFP-15CL) induced aberrant strands in MDCK II cells, even though claudin-15 has the ZO-binding motif. Morphometric analysis by freeze-fracture electron microscopy revealed that the mean number of apical TJ strands increased by 47% in EGFP-1CL-expressing cells, 21% in EGFP-15CL-expressing cells, and 28% in 15CL-expressing cells. The number of free-ended apical strands increased remarkably in EGFP-15CL- and 15CL-expressing cells, but not in EGFP-1CL-expressing cells. When MDCK cells expressing EGFP-1CL, EGFP-15CL or 15CL were co-cultured with parent MDCK cells, which express claudin-1 but not claudin-15, EGFP-15CL and 15CL could not be concentrated at the apical junctional region between the heterotypic cells, though EGFP-1CL could. These results suggest that not only binding to ZO-1, but also head-to-head compatibility between claudin species, is involved in organizing claudin proteins at the apical junctional region.
Collapse
Affiliation(s)
- Akihito Sengoku
- Department of Developmental Molecular Anatomy, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, 812-8582 Fukuoka, Japan.
| | | | | |
Collapse
|
156
|
Zhao S, Gu Y, Lewis DF, Wang Y. Predominant basal directional release of thromboxane, but not prostacyclin, by placental trophoblasts from normal and preeclamptic pregnancies. Placenta 2007; 29:81-8. [PMID: 17936899 DOI: 10.1016/j.placenta.2007.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2007] [Revised: 08/30/2007] [Accepted: 08/30/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To investigate apical and basal releases of thromboxane (TX) and prostacyclin (PGI2) by trophoblasts (TCs) from normal and preeclamptic (PE) placentas. METHODS TCs isolated from normal and PE placentas were incubated in cell culture inserts for 48h. Medium from the upper (apical) and the lower (basal) chambers were then collected separately and measured for TX and PGI2 by their stable metabolites of TXB2 and 6-keto PGF1alpha by ELISA. Apical and basal releases of TX and PGI were also examined with apical exposure of TCs to arachidonic acid (AA)+/-aspirin at different concentrations. Villous tissue expressions for PGI synthase, TX synthase and TX (TP) receptor were examined by immunohistochemistry. RESULTS (1) TXB2, but not 6-keto PGF1alpha, concentrations were significantly higher in the lower than in the upper chambers with both normal and PE TCs (p<0.01); (2) apical exposure of TCs to AA resulted in a significant increase in TX release towards both the upper and the lower chambers in normal TCs (p<0.01), but only a significant increase in the upper chamber in PE TCs (p<0.01); (3) aspirin could attenuate AA-induced TX release both in the upper and the lower chambers in normal, but not in PE, TCs (p<0.01), respectively; (4) there were no differences in 6-keto PGF1alpha productions both in normal and PE TCs treated with AA+/-aspirin; (5) intense staining of TX synthase and TP receptor was seen in syncytiotrophoblast layer, villous core vessels and stromal cells in preeclamptic placental tissue sections. CONCLUSION Predominant basal release of TX together with intense staining of TX synthase and TP receptor in trophoblasts, stromal cells and villous core vessels are found in placentas from PE. We speculate if predominant basal release of TX by TCs occurs in vivo as we found in our in vitro culture condition, basal released TX may play a significant role in increased placental vasoconstriction such as in PE.
Collapse
Affiliation(s)
- S Zhao
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | | | | | | |
Collapse
|
157
|
Fields MJ, Fields PA. Morphological characteristics of the bovine corpus luteum during the estrous cycle and pregnancy. Theriogenology 2007; 45:1295-325. [PMID: 16727884 DOI: 10.1016/0093-691x(96)00099-4] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/1995] [Accepted: 03/05/1996] [Indexed: 11/16/2022]
Abstract
The corpus luteum, one of the biological clocks of the estrous cycle and pregnancy, is known foremost for its production of progesterone that blocks the pituitary release of gonadotropins and prepares the uterus for a pregnancy. The cellular sources of this progesterone are the steroidogenic small and large luteal cells. Other luteal cells that are not steroidogenic, but are believed to have an important role in the function of this gland are the fibroblast, macrophages and endothelial cells. The most prominent luteal cell is the large steroidogenic cell characterized by an abundance of smooth endoplasmic reticulum and densely packed spherical mitochondria that are indicative of its contribution to most of the circulating progesterone believed to be constitutively secreted and not under the control of LH. Other distinguishing features of the large luteal cell are the presence of rough endoplasmic reticulum, prominent Golgi, and secretory granules that are indicative of endocrine cells. This cell undergoes dynamic changes across the estrous cycle and pregnancy, believed to reflect a change in progesterone and protein secretion that will eventually influence a successful pregnancy or another ovulation if pregnancy fails. The morphological characteristics of the bovine luteal cells are the focus of this review.
Collapse
Affiliation(s)
- M J Fields
- Department of Animal Science, University of Florida, Gainesville, FL 32611-0910, USA
| | | |
Collapse
|
158
|
Chen WL, Lin CT, Lo HF, Lee JW, Tu IH, Hu FR. The role of protein tyrosine phosphorylation in the cell-cell interactions, junctional permeability and cell cycle control in post-confluent bovine corneal endothelial cells. Exp Eye Res 2007; 85:259-69. [PMID: 17624326 DOI: 10.1016/j.exer.2007.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Revised: 04/14/2007] [Accepted: 04/25/2007] [Indexed: 12/31/2022]
Abstract
Cell-cell interaction, junctional permeability and contact inhibition are important mechanisms that allow corneal endothelial cells to maintain stable corneal hydration status and also keep these cells in non-proliferative status. Protein tyrosine phosphatases (PTPs) are well known to play an important role in regulating cell-cell contacts, growth and differentiation. Inhibition of PTPs activity by a general PTP inhibitor has been proved to trigger post-confluent rat corneal endothelial cells to reenter cell cycles. In this study, we aimed to evaluate whether protein tyrosine phosphorylation is involved in cell-cell interactions, junctional permeability and cell cycle control in post-confluent, contact inhibited bovine corneal endothelial cells. Confluent cultures of bovine corneal endothelial cells were treated with different concentrations of general phosphatase inhibitor, sodium orthovanadate (vanadate) for several different durations. Protein tyrosine phosphorylation was confirmed by Western blot analysis. Immunocytochemistry was used to evaluate the effect of vanadate on adherens-type junctional proteins by staining of p120, N-cadherin and alpha-catenin. Paracelluar permeability was evaluated by transepithelial electric resistance. The effect of vanadate on cell cycle progression was confirmed by immunostaining of Ki67. Western blot analysis was used to evaluate the expression level of cell-cycle-associated proteins, including PCNA, cyclin D1, cyclin E and cyclin A. Time-dependent effects of vanadate on protein tyrosine phosphorylation were confirmed by Western blot analysis. ICC demonstrated the effect of vanadate on the disruption of p120, N-cadherin and alpha-catenin. Time- and dose-effects of vanadate on the severity of p120 disruption were also found. TER demonstrated the time- and dose-effect of vanadte on paracellular permeability. Although cell-cell junctions can be broken through by vanadate, no significant increase of Ki67 positive cells was found among the control group and all groups with different concentrations/durations of vanadate treatment. Western blot also showed no change of PCNA, cyclin D1, cyclin E and cyclin A after treatment with vanadate. In conclusion, protein tyrosine phosphatase inhibition can induce time-dependent release of cell-cell contacts and increase transepithelial permeability in post-confluent cultures of bovine corneal endothelial cells. However, such phenomenon is not enough to promoted cell cycle progression as seen in rat corneal endothelial cells.
Collapse
Affiliation(s)
- Wei-Li Chen
- Department of Ophthalmology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
159
|
Saez A, Ghibaudo M, Buguin A, Silberzan P, Ladoux B. Rigidity-driven growth and migration of epithelial cells on microstructured anisotropic substrates. Proc Natl Acad Sci U S A 2007; 104:8281-6. [PMID: 17488828 PMCID: PMC1895941 DOI: 10.1073/pnas.0702259104] [Citation(s) in RCA: 258] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The physical properties of the cellular environment are involved in regulating the formation and maintenance of tissues. In particular, substrate rigidity appears to be a key factor dictating cell response on culture surfaces. Here we study the behavior of epithelial cells cultured on microfabricated substrates engineered to exhibit an anisotropic stiffness. The substrate consists of a dense array of micropillars of oval cross-section, so that one direction is made stiffer than the other. We demonstrate how such an anisotropic rigidity can induce directional epithelial growth and guide cell migration along the direction of greatest rigidity. Regions of high tractional stress and large cellular deformations within the sheets of cells are concentrated at the edges, in particular at the two poles of the islands along their long axis, in correlation with the orientation of actin stress fibers and focal adhesions. By inducing scattering activity of epithelial cells, we show that isolated cells also migrate along the direction of greatest stiffness. Taken together, these findings show that the mechanical interactions of cells with their microenvironment can be tuned to engineer particular tissue properties.
Collapse
Affiliation(s)
- Alexandre Saez
- *Laboratoire Matière et Systèmes Complexes, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7057, Batiment Condorcet, Université Paris 7, 10, rue Alice Domon et Léonie Duquet, F-75205 Paris Cedex 13, France; and
| | - Marion Ghibaudo
- *Laboratoire Matière et Systèmes Complexes, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7057, Batiment Condorcet, Université Paris 7, 10, rue Alice Domon et Léonie Duquet, F-75205 Paris Cedex 13, France; and
| | - Axel Buguin
- Physico-Chimie Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 168, Institut Curie, 26, rue d'Ulm, F-75248 Paris Cedex 05, France
| | - Pascal Silberzan
- Physico-Chimie Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 168, Institut Curie, 26, rue d'Ulm, F-75248 Paris Cedex 05, France
| | - Benoît Ladoux
- *Laboratoire Matière et Systèmes Complexes, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7057, Batiment Condorcet, Université Paris 7, 10, rue Alice Domon et Léonie Duquet, F-75205 Paris Cedex 13, France; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
160
|
Chandramouly G, Abad PC, Knowles DW, Lelièvre SA. The control of tissue architecture over nuclear organization is crucial for epithelial cell fate. J Cell Sci 2007; 120:1596-606. [PMID: 17405811 DOI: 10.1242/jcs.03439] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The remodeling of nuclear organization during differentiation and the dramatic alteration of nuclear organization associated with cancer development are well documented. However, the importance of tissue architecture in the control of nuclear organization remains to be determined. Differentiation of mammary epithelial cells into functional tissue structures, in three-dimensional culture, is characterized by a specific tissue architecture (i.e. a basoapical polarity axis), cell cycle exit and maintenance of cell survival. Here we show that induction of partial differentiation (i.e. basal polarity only, cell cycle exit and cell survival) by epigenetic mechanisms in malignant breast cells is sufficient to restore features of differentiation-specific nuclear organization, including perinucleolar heterochromatin, large splicing factor speckles, and distinct nuclear mitotic apparatus protein (NuMA) foci. Upon alteration of nuclear organization using an antibody against NuMA, differentiated non-neoplastic cells undergo apoptosis, whereas partially differentiated malignant cells enter the cell cycle. Non-neoplastic cells cultured under conditions that prevent the establishment of apical polarity also enter the cell cycle upon NuMA antibody treatment. These findings demonstrate that the differentiation status rather than the non-neoplastic or neoplastic origin of cells controls nuclear organization and suggest a link between nuclear organization and epigenetic mechanisms dictated by tissue architecture for the control of cell behavior.
Collapse
Affiliation(s)
- Gurushankar Chandramouly
- Department of Basic Medical Sciences and Cancer Center, Purdue University, 625 Harrison Street, West Lafayette, IN 47907-2026, USA
| | | | | | | |
Collapse
|
161
|
Fink C, Weigel R, Hembes T, Lauke-Wettwer H, Kliesch S, Bergmann M, Brehm RH. Altered expression of ZO-1 and ZO-2 in Sertoli cells and loss of blood-testis barrier integrity in testicular carcinoma in situ. Neoplasia 2007; 8:1019-27. [PMID: 17217619 PMCID: PMC1783719 DOI: 10.1593/neo.06559] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Carcinoma in situ (CIS) is the noninvasive precursor of most human testicular germ cell tumors. In normal seminiferous epithelium, specialized tight junctions between Sertoli cells constitute the major component of the blood-testis barrier. Sertoli cells associated with CIS exhibit impaired maturation status, but their functional significance remains unknown. The aim was to determine whether the blood-testis barrier is morphologically and/or functionally altered. We investigated the expression and distribution pattern of the tight junction proteins zonula occludens (ZO) 1 and 2 in normal seminiferous tubules compared to tubules showing CIS. In normal tubules, ZO-1 and ZO-2 immunostaining was observed at the blood-testis barrier region of adjacent Sertoli cells. Within CIS tubules, ZO-1 and ZO-2 immunoreactivity was reduced at the blood-testis barrier region, but spread to stain the Sertoli cell cytoplasm. Western blot analysis confirmed ZO-1 and ZO-2, and their respective mRNA were shown by RT-PCR. Additionally, we assessed the functional integrity of the blood-testis barrier by lanthanum tracer study. Lanthanum permeated tight junctions in CIS tubules, indicating disruption of the blood-testis barrier. In conclusion, Sertoli cells associated with CIS show an altered distribution of ZO-1 and ZO-2 and lose their blood-testis barrier function.
Collapse
Affiliation(s)
- Cornelia Fink
- Institute of Veterinary Anatomy, Histology, and Embryology, University of Giessen, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
162
|
Orsi NM, Reischl JB. Mammalian embryo co-culture: Trials and tribulations of a misunderstood method. Theriogenology 2007; 67:441-58. [PMID: 17118433 DOI: 10.1016/j.theriogenology.2006.10.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Revised: 10/17/2006] [Accepted: 10/19/2006] [Indexed: 11/17/2022]
Abstract
Embryo-somatic cell co-culture was devised over 40 years ago in an attempt to improve the development and viability of mammalian preimplantation embryos generated and cultured in vitro. While initial endeavours were successful in this respect, other studies soon highlighted a number of significant long-term detrimental impacts of this approach. Surprisingly little is known about the mechanisms underlying the beneficial effects of co-culture, although the production of embryotrophic compounds, modulation of nutrient profile, protection against culture-induced stress and/or toxin clearance are all contenders. The extent to which the inadvertent exposure of embryos to serum accounts for many of these effects remains open to question. Although the popularity of somatic cell co-culture has recently declined in favour of the use of sequential media due to concerns associated with its risk of disease transmission and long-term sequelae, we argue that complete dismissal of this technique is ill advised, given that our limited understanding of basic somatic cell interactions has prevented us from fully exploiting its potential. In this respect, there is some merit in focussing future research strategies based on reconstructed maternal tract tissue. Although the use of co-culture in clinical practice is unacceptable and its implementation in domestic species for commercial purposes should be viewed with diffidence, this technique can still provide a wealth of information on the development of novel, more physiological embryo in vitro culture systems. The proviso for acquiring such information is to gain a fuller understanding of the culture requirements/biochemistry of somatic cells and their interaction with the early conceptus.
Collapse
Affiliation(s)
- Nicolas M Orsi
- Perinatal Research Group, Section of Pathology & Tumour Biology, Leeds Institute of Molecular Medicine, Level 4 Wellcome Trust Brenner Building, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK.
| | | |
Collapse
|
163
|
Abstract
Electron microscopy (EM) has been a central tool in delineating the subcellular organization and function of the eukaryotic cell. It has provided valuable information on the organization of the Golgi complex; the polarized distribution of proteins on the plasma membrane; and fundamental insights into the essential structure and function of mitochondria beginning with the first EM observations of Claude and Fullam on isolated mitochondria in 1944. Most significant for this volume is the contribution immunoelectron microscopy (IEM) has made in the study of mitochondrial dynamics and in demonstrating the localizations of key mitochondrial proteins in yeast, including, though not limited to, Dnm1p, Fiz1p, and Mgm1p. This chapter is not intended to provide a comprehensive review of all EM and IEM methods as there are a number of excellent books and reviews already available on these topics. Rather, this chapter provides detailed protocols of conventional EM and IEM methods successfully utilized in our center for the examination and analysis of mitochondria in yeast and mammalian cells.
Collapse
|
164
|
Hilborn J, Bjursten LM. A new and evolving paradigm for biocompatibility. J Tissue Eng Regen Med 2007; 1:110-9. [DOI: 10.1002/term.4] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
165
|
Gizard F, Robillard R, Gross B, Barbier O, Révillion F, Peyrat JP, Torpier G, Hum DW, Staels B. TReP-132 is a novel progesterone receptor coactivator required for the inhibition of breast cancer cell growth and enhancement of differentiation by progesterone. Mol Cell Biol 2006; 26:7632-44. [PMID: 17015480 PMCID: PMC1636875 DOI: 10.1128/mcb.00326-06] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The sex steroid progesterone is essential for the proliferation and differentiation of the mammary gland epithelium during pregnancy. In relation to this, in vitro studies using breast carcinoma T47D cells have demonstrated a biphasic progesterone response, consisting of an initial proliferative burst followed by a sustained growth arrest. However, the transcriptional factors acting with the progesterone receptor (PR) to mediate the progesterone effects on mammary cell growth and differentiation remain to be determined. Recently, it has been demonstrated that the transcriptional regulating protein of 132 kDa (TReP-132), initially identified as a regulator of steroidogenesis, is also a cell growth suppressor. Similar to progesterone-bound PR, TReP-132 acts by inducing the gene expression of the G1 cyclin-dependent kinase inhibitors p21WAF1/Cip1 (p21) and p27Kip1 (p27). The putative interaction between TReP-132 and progesterone pathways in mammary cells was therefore analyzed in the present study. Our results show that TReP-132 interacts in vitro and in T47D cells with progesterone-activated PR. TReP-132 synergizes with progesterone-bound PR to trans activate the p21 and p27 gene promoters at proximal Sp1-binding sites. Moreover, TReP-132 overexpression and knockdown, respectively, increased or prevented the induction of p21 and p27 gene expression by progesterone. As a consequence, TReP-132 knockdown also resulted in the loss of the inhibitory effects of progesterone on pRB phosphorylation, G1/S cell cycle progression, and cell proliferation. Furthermore, the knockdown of TReP-132 expression also prevented the induction of both early and terminal markers of breast cell differentiation which had been previously identified as progesterone target genes. As well, the progesterone-induced accumulation of lipid vacuoles was inhibited in the TReP-132-depleted cells. Finally, TReP-132 gene expression levels increased following progesterone treatment, indicating the existence of a positive auto-regulatory loop between PR and TReP-132. Taken together, these data identify TReP-132 as a coactivator of PR mediating the growth-inhibitory and differentiation effects of progesterone on breast cancer cells.
Collapse
Affiliation(s)
- Florence Gizard
- INSERM U545, Institut Pasteur de Lille, 1 rue Calmette, BP 245, 59019 Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Hayashida Y, Urata Y, Muroi E, Kono T, Miyata Y, Nomata K, Kanetake H, Kondo T, Ihara Y. Calreticulin Represses E-cadherin Gene Expression in Madin-Darby Canine Kidney Cells via Slug. J Biol Chem 2006; 281:32469-84. [PMID: 16943193 DOI: 10.1074/jbc.m607240200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calreticulin (CRT) is a multifunctional Ca(2+)-binding molecular chaperone in the endoplasmic reticulum. In mammals, the expression level of CRT differs markedly in a variety of organs and tissues, suggesting that CRT plays a specific role in each cell type. In the present study, we focused on CRT functions in the kidney, where overall expression of CRT is quite low, and established CRT-overexpressing kidney epithelial cell-derived Madin-Darby canine kidney cells by gene transfection. We demonstrated that, in CRT-overexpressing cells, the morphology was apparently changed, and the original polarized epithelial cell phenotype was destroyed. Furthermore, CRT-overexpressing cells showed enhanced migration through Matrigel-coated Boyden chamber wells, compared with controls. E-cadherin expression was significantly suppressed at the protein and transcriptional levels in CRT-overexpressing cells compared with controls. On the other hand, the expression of mesenchymal protein markers, such as N-cadherin and fibronectin, was up-regulated. We also found that the expression of Slug, a repressor of the E-cadherin promoter, was up-regulated by overexpression of CRT through altered Ca(2+) homeostasis, and this led to enhanced binding of Slug to the E-box element in the E-cadherin promoter. Thus, we conclude that CRT regulates the epithelial-mesenchymal transition-like change of cellular phenotype by modulating the Slug/E-cadherin pathway through altered Ca(2+) homeostasis in cells, suggesting a novel function of CRT in cell-cell interaction of epithelial cells.
Collapse
Affiliation(s)
- Yasushi Hayashida
- Department of Biochemistry and Molecular Biology in Disease, Atomic Bomb Disease Institute, and Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Komiya S, Shimizu M, Ikenouchi J, Yonemura S, Matsui T, Fukunaga Y, Liu H, Endo F, Tsukita S, Nagafuchi A. Apical membrane and junctional complex formation during simple epithelial cell differentiation of F9 cells. Genes Cells 2006; 10:1065-80. [PMID: 16236135 DOI: 10.1111/j.1365-2443.2005.00899.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epithelium formation is a common event in animal morphogenesis. It has been reported that F9 cells differentiate into visceral endoderm-like epithelial cells when cell aggregates are cultured in the presence of retinoic acid. The present investigation set out to determine whether this in vitro model could be used under monolayer culture conditions, which is suitable for a detailed analysis of epithelial differentiation. We performed comparative gene expression analyses of F9 cells grown under aggregate and monolayer culture conditions prior to and following treatment with retinoic acid. Under these conditions, induction in the expression of differentiation marker genes was confirmed, even in monolayer cultures. Junctional complex and apical membrane formation, both of which are characteristic of epithelial cells, were also observed under monolayer culture conditions. Because of the merit of monolayer culture condition, we found that apical membrane and junctional complex formation are strictly regulated during epithelial differentiation. It was also revealed that F9 cells differentiated into epithelial cells predominantly on the fourth and fifth day following retinoic acid induction. These results showed that a monolayer culture of F9 cells represents a viable in vitro model that can be employed to elucidate mechanisms pertaining to epithelium formation.
Collapse
Affiliation(s)
- Satoshi Komiya
- Division of Cellular Interactions, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Abstract
Ten years ago, par-1 and par-3 were cloned as two of the six par genes essential for the asymmetric division of the Caenorhabditis elegans zygote. PAR-1 is a protein kinase, whereas PAR-3 is a PDZ-domain-containing scaffold protein. Work over the past decade has shown that they are part of an evolutionarily conserved PAR-aPKC system involved in cell polarity in various biological contexts. Recent progress has illustrated the common principle that the PAR-aPKC system is the molecular machinery that converts initial polarity cues in the establishment of complementary membrane domains along the polarity axis. In most cases, this is achieved by mutually antagonistic interactions between the aPKC-PAR-3-PAR-6 complex and PAR-1 or PAR2 located opposite. However, accumulating evidence has also revealed that mechanisms by which the asymmetrically localized components of the PAR-aPKC system are linked with other cellular machinery for developing polarity are divergent depending on the cell type.
Collapse
Affiliation(s)
- Atsushi Suzuki
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan.
| | | |
Collapse
|
169
|
Jakob V, Schreiner A, Tikkanen R, Starzinski-Powitz A. Targeting of transmembrane protein shrew-1 to adherens junctions is controlled by cytoplasmic sorting motifs. Mol Biol Cell 2006; 17:3397-408. [PMID: 16707570 PMCID: PMC1525240 DOI: 10.1091/mbc.e05-11-1034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We recently identified transmembrane protein shrew-1 and showed that it is able to target to adherens junctions in polarized epithelial cells. This suggested shrew-1 possesses specific basolateral sorting motifs, which we analyzed by mutational analysis. Systematic mutation of amino acids in putative sorting signals in the cytoplasmic domain of shrew-1 revealed three tyrosines and a dileucine motif necessary for basolateral sorting. Substitution of these amino acids leads to apical localization of shrew-1. By applying tannic acid to either the apical or basolateral part of polarized epithelial cells, thereby blocking vesicle fusion with the plasma membrane, we obtained evidence that the apically localized mutants were primarily targeted to the basolateral membrane and were then redistributed to the apical domain. Further support for a postendocytic sorting mechanism of shrew-1 was obtained by demonstrating that mu1B, a subunit of the epithelial cell-specific adaptor complex AP-1B, interacts with shrew-1. In conclusion, our data provide evidence for a scenario where shrew-1 is primarily delivered to the basolateral membrane by a so far unknown mechanism. Once there, adaptor protein complex AP-1B is involved in retaining shrew-1 at the basolateral membrane by postendocytic sorting mechanisms.
Collapse
Affiliation(s)
- Viktor Jakob
- *Institute of Cell Biology and Neuroscience, Johann-Wolfgang Goethe University of Frankfurt, D-60323 Frankfurt am Main, Germany; and
| | - Alexander Schreiner
- *Institute of Cell Biology and Neuroscience, Johann-Wolfgang Goethe University of Frankfurt, D-60323 Frankfurt am Main, Germany; and
| | - Ritva Tikkanen
- Institute of Biochemistry II, University Clinic of Frankfurt, D-60590 Frankfurt am Main, Germany
| | - Anna Starzinski-Powitz
- *Institute of Cell Biology and Neuroscience, Johann-Wolfgang Goethe University of Frankfurt, D-60323 Frankfurt am Main, Germany; and
| |
Collapse
|
170
|
Hirose T, Karasawa M, Sugitani Y, Fujisawa M, Akimoto K, Ohno S, Noda T. PAR3 is essential for cyst-mediated epicardial development by establishing apical cortical domains. Development 2006; 133:1389-98. [PMID: 16510507 DOI: 10.1242/dev.02294] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Epithelial cysts are one of the fundamental architectures for mammalian organogenesis. Although in vitro studies using cultured epithelial cells have revealed proteins required for cyst formation, the mechanisms that orchestrate the functions of these proteins in vivo remain to be clarified. We show that the targeted disruption of the mouse Par3 gene results in midgestational embryonic lethality with defective epicardial development. The epicardium is mainly derived from epicardial cysts and essential for cardiomyocyte proliferation during cardiac morphogenesis. PAR3-deficient epicardial progenitor (EPP) cells do not form cell cysts and show defects in the establishment of apical cortical domains, but not in basolateral domains. In PAR3-deficient EPP cells, the localizations of aPKC, PAR6β and ezrin to the apical cortical domains are disturbed. By contrast, ZO1 andα4/β1 integrins normally localize to cell-cell junctions and basal domains, respectively. Our observations indicate that EPP cell cyst formation requires PAR3 to interpret the polarity cues from cell-cell and cell-extracellular matrix interactions so that each EPP cell establishes apical cortical domains. These results also provide a clear example of the proper organization of epithelial tissues through the regulation of individual cell polarity.
Collapse
Affiliation(s)
- Tomonori Hirose
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Yokohama 236-0004, Japan
| | | | | | | | | | | | | |
Collapse
|
171
|
Brunham LR, Kruit JK, Iqbal J, Fievet C, Timmins JM, Pape TD, Coburn BA, Bissada N, Staels B, Groen AK, Hussain MM, Parks JS, Kuipers F, Hayden MR. Intestinal ABCA1 directly contributes to HDL biogenesis in vivo. J Clin Invest 2006; 116:1052-62. [PMID: 16543947 PMCID: PMC1401485 DOI: 10.1172/jci27352] [Citation(s) in RCA: 383] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 01/17/2006] [Indexed: 11/17/2022] Open
Abstract
Plasma HDL cholesterol levels are inversely related to risk for atherosclerosis. The ATP-binding cassette, subfamily A, member 1 (ABCA1) mediates the rate-controlling step in HDL particle formation, the assembly of free cholesterol and phospholipids with apoA-I. ABCA1 is expressed in many tissues; however, the physiological functions of ABCA1 in specific tissues and organs are still elusive. The liver is known to be the major source of plasma HDL, but it is likely that there are other important sites of HDL biogenesis. To assess the contribution of intestinal ABCA1 to plasma HDL levels in vivo, we generated mice that specifically lack ABCA1 in the intestine. Our results indicate that approximately 30% of the steady-state plasma HDL pool is contributed by intestinal ABCA1 in mice. In addition, our data suggest that HDL derived from intestinal ABCA1 is secreted directly into the circulation and that HDL in lymph is predominantly derived from the plasma compartment. These data establish a critical role for intestinal ABCA1 in plasma HDL biogenesis in vivo.
Collapse
Affiliation(s)
- Liam R. Brunham
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Janine K. Kruit
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jahangir Iqbal
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Catherine Fievet
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Jenelle M. Timmins
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Terry D. Pape
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Bryan A. Coburn
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Nagat Bissada
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Bart Staels
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Albert K. Groen
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - M. Mahmood Hussain
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - John S. Parks
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Folkert Kuipers
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Michael R. Hayden
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
Center for Liver, Digestive and Metabolic Diseases, Laboratory of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands.
Department of Anatomy and Cell Biology, State University of New York Downstate Medical Center, New York, New York, USA.
Institut Pasteur de Lille and Faculté de Pharmacie, Université de Lille, Lille, France.
Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
Department of Microbiology, University of British Columbia, Vancouver, British Columbia, Canada.
Department of Experimental Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
172
|
Füllekrug J, Shevchenko A, Shevchenko A, Simons K. Identification of glycosylated marker proteins of epithelial polarity in MDCK cells by homology driven proteomics. BMC BIOCHEMISTRY 2006; 7:8. [PMID: 16533391 PMCID: PMC1421407 DOI: 10.1186/1471-2091-7-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 03/13/2006] [Indexed: 11/24/2022]
Abstract
Background MDCK cells derived from canine kidney are an important experimental model system for investigating epithelial polarity in mammalian cells. Monoclonal antibodies against apical gp114 and basolateral p58 have served as important tools in these studies. However, the molecular identity of these membrane glycoproteins has not been known. Results We have identified the sialoglycoprotein gp114 as a dog homologue of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family. Gp114 was enriched from tissue culture cells by subcellular fractionation and immunoaffinity chromatography. The identification was based on tandem mass spectrometry and homology based proteomics. In addition, the p58 basolateral marker glycoprotein was found to be the β subunit of Na+K+-ATPase. Conclusion Gp114 has been characterized previously regarding glycosylation dependent trafficking and lipid raft association. The identification as a member of the canine CEACAM family will enable synergy between the fields of epithelial cell biology and other research areas. Our approach exemplifies how membrane proteins can be identified from species with unsequenced genomes by homology based proteomics. This approach is applicable to any model system.
Collapse
Affiliation(s)
- Joachim Füllekrug
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- University of Heidelberg, Internal Medicine IV, Molecular Cell Biology Group, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Anna Shevchenko
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrej Shevchenko
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Kai Simons
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
173
|
Abstract
Polarized epithelial cells are characterized by distinct plasma membrane domains and asymmetrical distribution of cell surface proteins and lipids. In vertebrates, tight junctions act as a fence between the apical and basolateral domains. Although many of the key components of the polarity machinery have been identified, their functions in cell polarization and junction formation remain to be determined. With the rapid improvement of the RNA interference (RNAi) technique, it is now possible to silence the expression of these polarity proteins in mammalian cells and to systematically analyze their distinct roles in orchestrating the polarization program. Here we describe approaches to achieve specific gene suppression in MDCK cells, a well-established cell culture model of canine kidney cells. We discuss the potential challenges and problems associated with the RNAi technique and describe basic protocols for suppressing gene expression using a vector-based short hairpin RNA (shRNA) expression system coupled with nucleofection.
Collapse
Affiliation(s)
- Xinyu Chen
- Center for Cell Signalling, University of Virginia School of Medicine, Charlottesville, USA
| | | |
Collapse
|
174
|
|
175
|
Influence of Porphyromonas gingivalis on the Expression of Cell Adhesion Molecules and Apoptosis in Human Gingival Epithelial Cells. J Oral Biosci 2006. [DOI: 10.1016/s1349-0079(06)80023-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
176
|
Schepers MSJ, van Ballegooijen ES, Bangma CH, Verkoelen CF. Oxalate is toxic to renal tubular cells only at supraphysiologic concentrations. Kidney Int 2005; 68:1660-9. [PMID: 16164643 DOI: 10.1111/j.1523-1755.2005.00576.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Oxalate-induced tissue damage may play an initiating role in the pathophysiology of calcium oxalate nephrolithiasis. The concentration of oxalate is higher in the renal collecting ducts ( approximately 0.1 to 0.5 mmol/L) than in the proximal tubule ( approximately 0.002 to 0.1 mmol/L). In the present investigation, we studied the damaging effect of oxalate to renal proximal and collecting tubule cells in culture. METHODS Studies were performed with the renal proximal tubular cell lines, LLC-PK1 and Madin Darby canine kidney II (MDCK-II), and the renal collecting duct cell lines, rat renal cortical collecting duct (RCCD1) and MDCK-I. Confluent monolayers cultured on permeable growth substrates in a two-compartment culture system were apically exposed for 24 hours to relatively low (0.2, 0.5, and 1.0 mmol/L) and high (5 and 10 mmol/L) oxalate concentrations, after which several cellular responses were studied, including monolayer morphology (confocal microscopy), transepithelial electrical resistances (TER), prostaglandin E(2) (PGE(2)) secretion, lactate dehydrogenase (LDH) release, DNA synthesis ([(3)H]-thymidine incorporation), total cell numbers, reactive oxygen species (H(2)O(2)) generation, apoptotic (annexin V and DNA fragmentation), and necrotic (propidium iodide influx) cell death. RESULTS Visible morphologic alterations were observed only at high oxalate concentrations. TER was concentration-dependently decreased by high, but not by low, oxalate. Elevated levels of PGE(2), LDH, and H(2)O(2) were measured in both cell types after exposure to high, but not to low oxalate. Exposure to high oxalate resulted in elevated levels of DNA synthesis with decreasing total cell numbers. High, but not low, oxalate induced necrotic cell death without signs of programmed cell death. CONCLUSION This study shows that oxalate is toxic to renal tubular cells, but only at supraphysiologic concentrations.
Collapse
Affiliation(s)
- Marieke S J Schepers
- Department of Urology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
177
|
Abstract
The Na,K-adenosine triphosphatase (ATPase), or sodium pump, has been well studied for its role in the regulation of ion homeostasis in mammalian cells. Recent studies suggest that Na,K-ATPase might have multiple functions such as a role in the regulation of tight junction structure and function, induction of polarity, regulation of actin dynamics, control of cell movement, and cell signaling. These functions appear to be modulated by Na,K-ATPase enzyme activity as well as protein-protein interactions of the alpha and beta subunits. In this review we attempt to differentiate functions associated with enzyme activity and subunit interactions. In addition, the consequence of impaired Na,K-ATPase function or reduced subunit expression levels in kidney diseases such as cancer, tubulointerstitial fibrosis, and ischemic nephropathy are discussed.
Collapse
Affiliation(s)
- Sigrid A Rajasekaran
- Department of Pathology and Laboratory Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
178
|
Gill GA, Buda A, Moorghen M, Dettmar PW, Pignatelli M. Characterisation of adherens and tight junctional molecules in normal animal larynx; determining a suitable model for studying molecular abnormalities in human laryngopharyngeal reflux. J Clin Pathol 2005; 58:1265-70. [PMID: 16311345 PMCID: PMC1770809 DOI: 10.1136/jcp.2004.016972] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2004] [Indexed: 11/04/2022]
Abstract
BACKGROUND The disruption of intercellular junctions in the larynx is a pathological feature of laryngopharyngeal reflux (LPR). Good experimental models are necessary to gain greater insight into the molecular mechanisms and alterations that result from abnormal exposure of the laryngeal epithelium to acid refluxate. AIMS To characterise laryngeal tissues from different species to determine the most suitable for use in experimental studies of LPR. METHODS Human and non-human laryngeal tissues (mouse, rat, guinea pig, porcine, and rabbit) were studied. Histological characterisation was performed by light microscopy. The expression and subcellular localisation of adherens junctional molecules (E-cadherin and beta catenin) was evaluated by immunohistochemistry, and tight junction molecules (occludin and zonula occludens 1 (ZO-1)) by western blotting. The ultrastructural features of porcine and human tissue were assessed by electron microscopy. RESULTS Porcine tissue revealed both respiratory-type and stratified squamous epithelium, as seen in the human larynx. The expression and subcellular localisation of the E-cadherin-catenin complex was detected in all species except mouse and rat. The pattern of ZO-1 and occludin expression was preserved in all species. CONCLUSION The expression of intercellular junctional complexes in porcine epithelium is similar to that seen in humans. These results confirm the suitability of these species to study molecular mechanisms of LPR in an experimental system.
Collapse
Affiliation(s)
- G A Gill
- Division of Histopathology, Department of Pathology and Microbiology, School of Medical Sciences and Bristol Royal Infirmary, University of Bristol, Bristol BS2 8HW, UK
| | | | | | | | | |
Collapse
|
179
|
Vandewalle C, Comijn J, De Craene B, Vermassen P, Bruyneel E, Andersen H, Tulchinsky E, Van Roy F, Berx G. SIP1/ZEB2 induces EMT by repressing genes of different epithelial cell-cell junctions. Nucleic Acids Res 2005; 33:6566-78. [PMID: 16314317 PMCID: PMC1298926 DOI: 10.1093/nar/gki965] [Citation(s) in RCA: 422] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
SIP1/ZEB2 is a member of the deltaEF-1 family of two-handed zinc finger nuclear factors. The expression of these transcription factors is associated with epithelial mesenchymal transitions (EMT) during development. SIP1 is also expressed in some breast cancer cell lines and was detected in intestinal gastric carcinomas, where its expression is inversely correlated with that of E-cadherin. Here, we show that expression of SIP1 in human epithelial cells results in a clear morphological change from an epithelial to a mesenchymal phenotype. Induction of this epithelial dedifferentiation was accompanied by repression of several cell junctional proteins, with concomitant repression of their mRNA levels. Besides E-cadherin, other genes coding for crucial proteins of tight junctions, desmosomes and gap junctions were found to be transcriptionally regulated by the transcriptional repressor SIP1. Moreover, study of the promoter regions of selected genes by luciferase reporter assays and chromatin immunoprecipitation shows that repression is directly mediated by SIP1. These data indicate that, during epithelial dedifferentiation, SIP1 represses in a coordinated manner the transcription of genes coding for junctional proteins contributing to the dedifferentiated state; this repression occurs by a general mechanism mediated by Smad Interacting Protein 1 (SIP1)-binding sites.
Collapse
Affiliation(s)
| | | | | | | | - Erik Bruyneel
- Laboratory of Experimental Cancerology, Department of Radiotherapy and Nuclear Medicine, Ghent UniversityBelgium
| | | | - Eugene Tulchinsky
- Department of Cancer Studies and Molecular Medicine, University of LeicesterUK
| | - Frans Van Roy
- Molecular Cell Biology Unit, Department for Molecular Biomedical Research, VIB-Ghent UniversityBelgium
| | - Geert Berx
- To whom correspondence should be addressed at Department for Molecular Biomedical Research, VIB-Ghent University, Technologiepark 927, 9052 Ghent (Zwijnaarde), Belgium. Tel: +32.0 9.3313740; Fax: +32.0 9.3313609;
| |
Collapse
|
180
|
Sun T, Zhu J, Yang X, Wang S. Growth of miniature pig parotid cells on biomaterials in vitro. Arch Oral Biol 2005; 51:351-8. [PMID: 16274660 DOI: 10.1016/j.archoralbio.2005.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2005] [Revised: 09/05/2005] [Accepted: 10/04/2005] [Indexed: 10/25/2022]
Abstract
Both Sjögren's syndrome and therapeutic irradiation for head and neck cancer lead to irreversible damage of the parenchyma of the salivary glands. This report describes an attempt to grow miniature pig (minipig) parotid gland cells on artificial films and tubular scaffolds with the ultimate intention of developing bio-engineered replacement tissue. Minipig parotid cells were isolated and cultured. The growth and structural and physiological features of the cells which were cultured on films and porous tubular scaffolds made from poly(ethylene glycol)-terephthalate (PEGT)/poly(butylene terephthalate) (PBT) were examined. By 9 days, the parotid cells on the films and the tubular scaffolds formed continuous monolayers. The secretory granules and nuclei of the cultured acinar cells remained polarised. Desmosomes, gap junctions and tight-like junctions were still present between the apical regions of adjacent cells. Amylase activity decreased during the culture period but was still evident in the medium after 10 days of culture. In conclusion, minipig parotid cells are well-maintained in vitro on both a flat surface and a three-dimensional (3D) scaffold. The addition of a Matrigel coating to the surface of synthetic materials aids cell growth and maintenance of a morphology that more closely resembles normal epithelium.
Collapse
Affiliation(s)
- Tao Sun
- Salivary Gland Disease Centre and the Molecular Laboratory for Gene Therapy, Faculty of Stomatology, Capital University of Medical Sciences, Beijing 100050, PR China
| | | | | | | |
Collapse
|
181
|
Abstract
Little is known about how the genotypic and molecular abnormalities associated with epithelial cancers actually contribute to the histological phenotypes observed in tumours in vivo. 3D epithelial culture systems are a valuable tool for modelling cancer genes and pathways in a structurally appropriate context. Here, we review the important features of epithelial structures grown in 3D basement membrane cultures, and how such models have been used to investigate the mechanisms associated with tumour initiation and progression.
Collapse
Affiliation(s)
- Jayanta Debnath
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
182
|
Linask KK, Manisastry S, Han M. Cross talk between cell-cell and cell-matrix adhesion signaling pathways during heart organogenesis: implications for cardiac birth defects. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2005; 11:200-8. [PMID: 16060972 DOI: 10.1017/s1431927605050440] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 10/27/2004] [Indexed: 05/03/2023]
Abstract
The anterior-posterior and dorsal-ventral progression of heart organogenesis is well illustrated by the patterning and activity of two members of different families of cell adhesion molecules: the calcium-dependent cadherins, specifically N-cadherin, and the extracellular matrix glycoproteins, fibronectin. N-cadherin by its binding to the intracellular molecule beta-catenin and fibronectin by its binding to integrins at focal adhesion sites, are involved in regulation of gene expression by their association with the cytoskeleton and through signal transduction pathways. The ventral precardiac mesoderm cells epithelialize and become stably committed by the activation of these cell-matrix and intracellular signaling transduction pathways. Cross talk between the adhesion signaling pathways initiates the characteristic phenotypic changes associated with cardiomyocyte differentiation: electrical activity and organization of myofibrils. The development of both organ form and function occurs within a short interval thereafter. Mutations in any of the interacting molecules, or environmental insults affecting either of these signaling pathways, can result in embryonic lethality or fetuses born with severe heart defects. As an example, we have defined that exposure of the embryo temporally to lithium during an early sensitive developmental period affects a canonical Wnt pathway leading to beta-catenin stabilization. Lithium exposure results in an anterior-posterior progression of severe cardiac defects.
Collapse
Affiliation(s)
- Kersti K Linask
- Department of Pediatrics, University of South Florida, College of Medicine, The Children's Research Institute, St. Petersburg, FL 33701, USA.
| | | | | |
Collapse
|
183
|
Le Bivic A. E-cadherin-mediated adhesion is not the founding event of epithelial cell polarity in Drosophila. Trends Cell Biol 2005; 15:237-40. [PMID: 15866027 DOI: 10.1016/j.tcb.2005.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Formation of a polarized epithelial layer is a fundamental step during the development of multicellular animals. This process involves the coordinated action of adhesion molecules, actin remodeling and spatial organization of membrane traffic. A recent article describes a new hierarchy for the development of epithelial polarity in the early Drosophila embryo. Bazooka, a Par-3 homolog, is properly localized in the absence of adherens junctions, indicating that the formation of epithelial junctions is not the founding event of epithelial polarization.
Collapse
Affiliation(s)
- André Le Bivic
- Developmental Biology Institute of Marseille, Faculté des Sciences de Luminy, UMR 6156, case 907, Marseille 13288, France.
| |
Collapse
|
184
|
Maruyama M, Kishimoto M, Ishida K, Watanabe Y, Nishikawa M, Masuda S, Sasaki R, Takakura Y. Cholesterol is required for the polarized secretion of erythropoietin in Madin-Darby canine kidney cells. Arch Biochem Biophys 2005; 438:174-81. [PMID: 15916748 DOI: 10.1016/j.abb.2005.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Revised: 04/11/2005] [Accepted: 04/11/2005] [Indexed: 11/21/2022]
Abstract
It has already been reported that stably expressed exogenous human wild-type EPO (wtEPO) is preferentially secreted to the apical side and one of the three N-linked carbohydrate chains critically acts as an apical sorting determinant in Madin-Darby canine kidney (MDCK) cells. It has been suggested that lipid rafts are involved in the apical sorting of membrane and secretory proteins. To investigate the involvement of lipid rafts in the apical sorting of wtEPO, we examined the effect of cholesterol depletion with methyl-beta-cyclodextrin on the secretion polarity of EPO and analyzed Triton X-100 insoluble cell extracts by sucrose density gradients centrifugation in MDCK cells. We found that wtEPO was shifted in non-polarized direction by cholesterol depletion. Most of the wtEPO was not detectable in the raft fractions by sucrose density gradients centrifugation analysis. These results indicate that apical secretion of EPO involves a cholesterol-dependent mechanism probably not involving lipid rafts.
Collapse
Affiliation(s)
- Masato Maruyama
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Japan
| | | | | | | | | | | | | | | |
Collapse
|
185
|
Hunt JL, Pollak MR, Denker BM. Cultured podocytes establish a size-selective barrier regulated by specific signaling pathways and demonstrate synchronized barrier assembly in a calcium switch model of junction formation. J Am Soc Nephrol 2005; 16:1593-602. [PMID: 15843471 DOI: 10.1681/asn.2004080679] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Podocytes form unique cell-cell junctions (slit diaphragms) that are central to glomerular selectivity, although regulation and mechanisms of slit diaphragm assembly are poorly understood. With the use of cultured podocytes, a paracellular permeability flux assay was established to characterize properties of the size-selective barrier. Paracellular flux of differentiated podocytes was measured using anionic fluorescent dextrans of 3, 10, 40, and 70 kD. Podocytes form a highly selective barrier with a 160-fold difference in flux from the 3-kD dextran (11 pmol/min) to the 70-kD dextran (0.06 pmol/min). Barrier development was dependent on podocyte differentiation and not affected by dextran charge. Puromycin, a known podocyte toxin, increased flux 250% in a dose-dependent manner without affecting cell viability. Screening with modulators of specific signaling pathways identified reversible increases in flux with Src tyrosine and Rho kinase inhibition. The calcium switch model of epithelial junction assembly was modified to determine whether podocytes regulate barrier assembly. When cultured in low calcium for 90 min, flux increased by 300% and consistently returned to baseline 24 to 48 h after switching to normal calcium. Similar to classical epithelial junctions, barrier recovery occurred in the presence of cyclohexamide, an inhibitor of protein synthesis. During the calcium switch, there were reversible changes in localization and detergent solubility of the slit diaphragm protein ZO-1 and alpha-actinin-4, whereas nephrin and podocin solubility were unchanged. Taken together, these findings demonstrate that cultured podocytes develop a selective size barrier that is regulated by specific signaling pathways, and similar to classical epithelial junctions, podocytes demonstrate synchronized assembly of the barrier.
Collapse
Affiliation(s)
- Jennifer L Hunt
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | |
Collapse
|
186
|
Rodriguez-Boulan E, Kreitzer G, Müsch A. Organization of vesicular trafficking in epithelia. Nat Rev Mol Cell Biol 2005; 6:233-47. [PMID: 15738988 DOI: 10.1038/nrm1593] [Citation(s) in RCA: 495] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Experiments using mammalian epithelial cell lines have elucidated biosynthetic and recycling pathways for apical and basolateral plasma-membrane proteins, and have identified components that guide apical and basolateral proteins along these pathways. These components include apical and basolateral sorting signals, adaptors for basolateral signals, and docking and fusion proteins for vesicular trafficking. Recent live-cell-imaging studies provide a real-time view of sorting processes in epithelial cells, including key roles for actin, microtubules and motors in the organization of post-Golgi trafficking.
Collapse
Affiliation(s)
- Enrique Rodriguez-Boulan
- Margaret Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, New York, New York 10021, USA.
| | | | | |
Collapse
|
187
|
Soulié P, Carrozzino F, Pepper MS, Strongin AY, Poupon MF, Montesano R. Membrane-type-1 matrix metalloproteinase confers tumorigenicity on nonmalignant epithelial cells. Oncogene 2005; 24:1689-97. [PMID: 15608664 DOI: 10.1038/sj.onc.1208360] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Overexpression of membrane-type-1 matrix metalloproteinase (MT1-MMP) in tumor cells has previously been shown to enhance tumor growth and metastasis. To establish if MT1-MMP is also able to confer tumorigenicity on nonmalignant epithelial cells, we transfected human MT1-MMP cDNA into Madin-Darby canine kidney (MDCK) cells expressing a tetracycline-repressible transactivator. Induction of MT1-MMP in the absence of doxycycline (Dox) was associated with activation of exogenous MMP-2 as well as with formation of large cysts and increased invasiveness in collagen matrices. Transfected cells were inoculated subcutaneously into two groups of nude mice, one of which received Dox to inhibit expression of MT1-MMP. Formation of tumor xenografts was observed in 11 of 17 mice maintained without Dox, but only in two of nine mice that received Dox (P<0.05). The xenografts were composed of tubular structures interspersed within a highly cellular stroma. The epithelial cells delimiting the lumen were polarized, as indicated by the basolateral distribution of Na,K-ATPase. Despite their differentiated appearance, the tumors lacked a well-defined boundary, and epithelial tubules invaded adjacent muscular layers. These results demonstrate that conditional expression of MT1-MMP in nonmalignant MDCK epithelial cells is by itself sufficient to drive formation of invasive tumors.
Collapse
Affiliation(s)
- Priscilla Soulié
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Rue Michel-Servet 1, CH-1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
188
|
Gautrey H, McConnell J, Hall J, Hesketh J. Polarised distribution of the RNA-binding protein Staufen in differentiated intestinal epithelial cells. FEBS Lett 2005; 579:2226-30. [PMID: 15811346 DOI: 10.1016/j.febslet.2005.02.074] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2004] [Revised: 01/21/2005] [Accepted: 02/08/2005] [Indexed: 11/22/2022]
Abstract
mRNA localisation, as a mechanism for directing localised protein synthesis, plays a vital role in the functioning of certain cells, such as neurons and oocytes. Potentially this mechanism may also occur in polarised intestinal epithelial cells. Here we show that Staufen1(55), a protein involved in mRNA localisation and transport, is asymmetrically distributed in differentiated Caco-2 intestinal epithelial cells and partly co-localised with calnexin, a marker of the endoplasmic reticulum. The localisation to the apical region of the cell indicates that Staufen may be involved in localisation of transcripts to this domain.
Collapse
Affiliation(s)
- Hannah Gautrey
- Institute for Cell and Molecular Biosciences, University of Newcastle, Newcastle-upon-Tyne NE1 7RU, UK
| | | | | | | |
Collapse
|
189
|
Harrell PC, McCawley LJ, Fingleton B, McIntyre JO, Matrisian LM. Proliferative effects of apical, but not basal, matrix metalloproteinase-7 activity in polarized MDCK cells. Exp Cell Res 2005; 303:308-20. [PMID: 15652345 DOI: 10.1016/j.yexcr.2004.09.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Revised: 09/27/2004] [Accepted: 09/28/2004] [Indexed: 10/26/2022]
Abstract
Matrix metalloproteinase-7 (MMP-7) is primarily expressed in glandular epithelium. Therefore, its mechanism of action may be influenced by its regulated vectorial release to either the apical and/or basolateral compartments, where it would act on its various substrates. To gain a better understanding of where MMP-7 is released in polarized epithelium, we have analyzed its pattern of secretion in polarized MDCK cells expressing stably transfected human MMP-7 (MDCK-MMP-7), and HCA-7 and Caco2 human colon cancer cell lines. In all cell lines, latent MMP-7 was secreted to both cellular compartments, but was 1.5- to 3-fold more abundant in the basolateral compartment as compared to the apical. However, studies in the MDCK system demonstrated that MMP-7 activity was 2-fold greater in the apical compartment of MDCK-MMP-7(HIGH)-polarized monolayers, which suggests the apical co-release of an MMP-7 activator. In functional assays, MMP-7 over-expression increased cell saturation density as a result of increased cell proliferation with no effect on apoptosis. Apical MMP-7 activity was shown to be responsible for the proliferative effect, which occurred, as demonstrated by media transfer experiments, through cleavage of an apical substrate and not through the generation of a soluble factor. Taken together, our findings demonstrate the importance of MMP-7 secretion in relation to its mechanism of action when expressed in a polarized epithelium.
Collapse
Affiliation(s)
- Permila C Harrell
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
190
|
Ideo H, Seko A, Yamashita K. Galectin-4 Binds to Sulfated Glycosphingolipids and Carcinoembryonic Antigen in Patches on the Cell Surface of Human Colon Adenocarcinoma Cells. J Biol Chem 2005; 280:4730-7. [PMID: 15546874 DOI: 10.1074/jbc.m410362200] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Galectin-4, a member of the galectin family, is expressed in the epithelium of the alimentary tract. It has two tandemly repeated carbohydrate recognition domains and specifically binds to an SO3- -->3Galbeta1-->3GalNAc pyranoside with high affinity (Ideo, H., Seko, A., Ohkura, T., Matta, K. L., and Yamashita, K. (2002) Glycobiology 12, 199-208). In this study, we found that galectin-4 binds to glycosphingolipids carrying 3-O-sulfated Gal residues, such as SB1a, SM3, SM4s, SB2, SM2a, and GM1, but not to glycosphingolipids with 3-O-sialylated Gal, such as sLc4Cer, snLc4Cer, GM3, GM2, and GM4, using both an enzyme-linked immunosorbent assay and a surface plasmon resonance assay. A confocal immunocytochemical assay showed that galectin-4 was colocalized with SB1a, GM1, and carcinoembryonic antigen (CEA) in the patches on the cell surface of human colon adenocarcinoma CCK-81 and LS174T cells. This localization was distinct from caveolin/VIP21 localization. Furthermore, immobilized galectin-4 promoted adhesion of CCK-81 cells through the sulfated glycosphingolipid, SB1a. CEA also bound to galectin-4 with KD value of 2 x 10(-8) m by surface plasmon resonance and coimmunoprecipitated with galectin-4 in LS174T cell lysates. These findings suggest that SB1a and CEA in the patches on the cell surface of human colon adenocarcinoma cells could be biologically important ligands for galectin-4.
Collapse
Affiliation(s)
- Hiroko Ideo
- Department of Biochemistry, Sasaki Institute, 2-2, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062 , Japan
| | | | | |
Collapse
|
191
|
Rüffer C, Gerke V. The C-terminal cytoplasmic tail of claudins 1 and 5 but not its PDZ-binding motif is required for apical localization at epithelial and endothelial tight junctions. Eur J Cell Biol 2005; 83:135-44. [PMID: 15260435 DOI: 10.1078/0171-9335-00366] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Claudins are a family of tetraspan transmembrane proteins that represent the major constituents of epithelial and endothelial tight junctions (TJs). They form TJ strands representing the major barrier regulating paracellular transport of solutes and water. Intracellularly, claudins are connected via a C-terminal PDZ-binding motif with several TJ-associated proteins containing PDZ domains. Although these interactions can provide a link to the actin cytoskeleton, they appear to be dispensable for the TJ localization of claudins. To identify TJ-targeting elements in the C-terminal cytoplasmic domains of the claudins 1 and 5, we generated a series of C-terminal deletion mutants and analyzed their distribution in polarized epithelial (MDCK) and endothelial (HMEC-1) cells. TJ localization was revealed by establishing an in vivo cross-linking approach that stabilized claudin-TJ interactions. We show that residues located C-terminal to the last transmembrane domain are required for the proper targeting to apical TJ.s. While claudin derivatives lacking only the very C-terminal PDZ-binding motif continue to localize to TJs, mutants lacking the entire C-terminal juxtamembrane sequence do not associate with TJs and accumulate in intracellular structures. This indicates that crucial determinants for stable TJ incorporation of claudins reside in a cytoplasmic C-terminal sequence which up to now has not been implicated in specific protein-protein interactions.
Collapse
Affiliation(s)
- Claas Rüffer
- Institute of Medical Biochcmistry, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | | |
Collapse
|
192
|
Faleiro-Rodrigues C, Macedo-Pinto I, Pereira D, Lopes CS. Prognostic value of E-cadherin immunoexpression in patients with primary ovarian carcinomas. Ann Oncol 2005; 15:1535-42. [PMID: 15367415 DOI: 10.1093/annonc/mdh387] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE To analyse the negative versus positive immunoexpression of E-cadherin in patients with primary ovarian carcinomas, and determine its significance in relation to clinicopathological features, overall and recurrence-free survival (RFS). PATIENTS AND METHODS The protein expression of E-cadherin was immunohistochemically evaluated in formalin-fixed, paraffin-embedded samples in 104 patients with primary ovarian carcinomas. The clinicopathological factors studied were age, FIGO staging, histological type, tumour differentiation, the appearance of the ovarian capsule, peritoneal implants and residual tumour after cytoreductive surgery. Overall survival and RFS were evaluated using the Kaplan-Meier method, and multivariate analysis was completed using the Cox regression model. RESULTS Of the 104 carcinomas, negative E-cadherin immunoexpression was observed in seven (7%) cases, and positive immunoexpression in 97 (93%). E-cadherin categorised into negative versus positive expression did not associate with any of the established clinicopathological parameters. However, negative E-cadherin expression significantly predicted a poorer overall survival when compared with positive expression (P=0.006). In the multivariate analyses, negative E-cadherin and the presence of residual tumour after cytoreductive surgery were independent prognostic factors for survival (P=0.014 and P=0.034, respectively). CONCLUSIONS The presence of residual tumour after primary cytoreductive surgery and negative E-cadherin expression seem to be useful markers in patients with ovarian carcinomas likely to have an unfavourable clinical outcome. The assessment of E-cadherin immunoreactivity may be a useful prognostic indicator in ovarian cancer, complementary to established prognostic factors.
Collapse
Affiliation(s)
- C Faleiro-Rodrigues
- Departments of Anatomy and Pathology and Medical Oncology, Portuguese Institute of Oncology of Francisco Gentil, Centro Regional do Norte, Porto, Portugal. cristinafaleiro@mailcom
| | | | | | | |
Collapse
|
193
|
Barwe SP, Anilkumar G, Moon SY, Zheng Y, Whitelegge JP, Rajasekaran SA, Rajasekaran AK. Novel role for Na,K-ATPase in phosphatidylinositol 3-kinase signaling and suppression of cell motility. Mol Biol Cell 2004; 16:1082-94. [PMID: 15616195 PMCID: PMC551475 DOI: 10.1091/mbc.e04-05-0427] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The Na,K-ATPase, consisting of alpha- and beta-subunits, regulates intracellular ion homeostasis. Recent studies have demonstrated that Na,K-ATPase also regulates epithelial cell tight junction structure and functions. Consistent with an important role in the regulation of epithelial cell structure, both Na,K-ATPase enzyme activity and subunit levels are altered in carcinoma. Previously, we have shown that repletion of Na,K-ATPase beta1-subunit (Na,K-beta) in highly motile Moloney sarcoma virus-transformed Madin-Darby canine kidney (MSV-MDCK) cells suppressed their motility. However, until now, the mechanism by which Na,K-beta reduces cell motility remained elusive. Here, we demonstrate that Na,K-beta localizes to lamellipodia and suppresses cell motility by a novel signaling mechanism involving a cross-talk between Na,K-ATPase alpha1-subunit (Na,K-alpha) and Na,K-beta with proteins involved in phosphatidylinositol 3-kinase (PI3-kinase) signaling pathway. We show that Na,K-alpha associates with the regulatory subunit of PI3-kinase and Na,K-beta binds to annexin II. These molecular interactions locally activate PI3-kinase at the lamellipodia and suppress cell motility in MSV-MDCK cells, independent of Na,K-ATPase ion transport activity. Thus, these results demonstrate a new role for Na,K-ATPase in regulating carcinoma cell motility.
Collapse
Affiliation(s)
- Sonali P Barwe
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
194
|
James PS, Hennessy C, Berge T, Jones R. Compartmentalisation of the sperm plasma membrane: a FRAP, FLIP and SPFI analysis of putative diffusion barriers on the sperm head. J Cell Sci 2004; 117:6485-95. [PMID: 15572407 DOI: 10.1242/jcs.01578] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spermatozoa are highly polarised cells with a compartmentalised distribution of lipids and proteins in their plasma membrane. It is not known how these compartments are stably maintained in what is essentially a fluid environment. In this investigation we have examined the hypothesis that intramembranous diffusion barriers selectively retain some components within compartments, while allowing free passage of others. A fluorescence loss in photobleaching analysis of the behaviour of the lipid reporter probe 1,1′-dihexadecyl-3,3,3′3′-tetramethyindocarbocyanine (DiIC16) on the head of boar spermatozoa revealed that it was freely diffusing between all three compartments (anterior acrosome, equatorial segment and postacrosome). Spermatozoa also contained rapidly diffusing particles of DiIC16 over the anterior acrosome and equatorial segment. These particles, ∼200 nm in diameter, were tracked in real time and their trajectories analysed by mean square displacement. Particle diffusion was essentially random over the anterior acrosome and equatorial segment but showed a periodicity in jump sizes and diffusion coefficients suggestive of microheterogeneities. Particles did not exchange between the equatorial segment and postacrosome, indicating a barrier at the junction between these two compartments. No barrier was detected between the equatorial segment and anterior acrosome. A model is proposed in which a molecular `filter' is present at the equatorial segment-postacrosomal boundary that allows free passage of single molecules but not molecular complexes. Passage of heterogeneous complexes, such as lipid rafts, requires disassembly and reassembly on either side of the filter.
Collapse
Affiliation(s)
- Peter S James
- Laboratory of Molecular Signalling, Babraham Institute, Cambridge, CB2 4AT, UK
| | | | | | | |
Collapse
|
195
|
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: 3.4] [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
|
196
|
Abstract
Components of paramyxoviruses are assembled at the plasma membrane of infected cells, and progeny viruses are formed by the budding process. Although the molecular mechanisms that drive budding (membrane curving and "pinching-off" reaction) are not well understood, the viral matrix (M) protein is thought to play a major role in the process. The M protein forms a dense layer tightly associated with the inner leaflet of the plasma membrane of infected cells. Expression of the M protein of some paramyxoviruses results in the formation and release of virus-like particles that contain the M protein; thus, in these viruses, the M protein alone can apparently trigger all steps required for the formation and release of virus-like particles. M also interacts specifically with viral envelope glycoproteins and nucleocapsids and is involved in directed transport of viral components to the budding site at the apical surface of polarized cells. In addition, protein-protein interactions between M and the cytoplasmic tail of viral glycoproteins and between M and the nucleocapsid affect the efficiency of virus production. The structural organization of the virion and the functions of the M protein clearly indicate that this protein orchestrates the budding of paramyxovirus.
Collapse
Affiliation(s)
- Toru Takimoto
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 672, Rochester, NY 14642, USA.
| | | |
Collapse
|
197
|
Campo C, Mason A, Maouyo D, Olsen O, Yoo D, Welling PA. Molecular mechanisms of membrane polarity in renal epithelial cells. Rev Physiol Biochem Pharmacol 2004; 153:47-99. [PMID: 15674648 DOI: 10.1007/s10254-004-0037-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Exciting discoveries in the last decade have cast light onto the fundamental mechanisms that underlie polarized trafficking in epithelial cells. It is now clear that epithelial cell membrane asymmetry is achieved by a combination of intracellular sorting operations, vectorial delivery mechanisms and plasmalemma-specific fusion and retention processes. Several well-defined signals that specify polarized segregation, sorting, or retention processes have, now, been described in a number of proteins. The intracellular machineries that decode and act on these signals are beginning to be described. In addition, the nature of the molecules that associate with intracellular trafficking vesicles to coordinate polarized delivery, tethering, docking, and fusion are also becoming understood. Combined with direct visualization of polarized sorting processes with new technologies in live-cell fluorescent microscopy, new and surprising insights into these once-elusive trafficking processes are emerging. Here we provide a review of these recent advances within an historically relevant context.
Collapse
Affiliation(s)
- C Campo
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
198
|
Wu J, Liu F, Nilsson A, Duan RD. Pancreatic trypsin cleaves intestinal alkaline sphingomyelinase from mucosa and enhances the sphingomyelinase activity. Am J Physiol Gastrointest Liver Physiol 2004; 287:G967-73. [PMID: 15205117 DOI: 10.1152/ajpgi.00190.2004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sphingomyelin (SM) hydrolysis in the gut has implications in colonic tumorigenesis and cholesterol absorption. It is triggered by intestinal alkaline sphingomyelinase (Alk-SMase) that is present in the intestinal mucosa and content. The mechanism by which the enzyme is released into the lumen is not clear. We studied whether trypsin can dissociate Alk-SMase from the mucosa and affect its activity. During luminal perfusion of rat intestine, addition of trypsin to the buffer increased Alk-SMase activity in the perfusate output by about threefold. Treating COS-7 cells transfected with Alk-SMase cDNA with trypsin increased the SMase activity in the medium and reduced that in the cell lysate dose dependently. The appearance of Alk-SMase in the perfusate and culture medium was confirmed by Western blot analysis. The effect of trypsin was blocked by trypsin inhibitor, and neither chymotrypsin nor elastase had a similar effect. We also expressed the full length and COOH-terminal truncated Alk-SMase in COS-7 cells and found that the activity of the full-length enzyme is mainly in the cells, whereas that of the truncated form is mainly in the medium. Both forms were active, but only the activity of the full-length Alk-SMase was enhanced by trypsin. By linking a poly-His tag to the constructed cDNA, we found that the first tryptic site Arg440 upstream of the signal anchor was attacked by trypsin. In conclusion, trypsin cleaves the Alk-SMase at the COOH terminal, releases it from mucosa, and meanwhile enhances its activity. The findings indicate a physiological role of trypsin in SM digestion.
Collapse
Affiliation(s)
- Jun Wu
- Gastroenterology Lab, Biomedical Center B11, Lund University, S-221 84 Lund, Sweden
| | | | | | | |
Collapse
|
199
|
Abstract
Mechanical forces play an important role in the organization, growth, maturation, and function of living tissues. At the cellular level, many of the biological responses to external forces originate at two types of specialized microscale structures: focal adhesions that link cells to their surrounding extracellular matrix and adherens junctions that link adjacent cells. Transmission of forces from outside the cell through cell-matrix and cell-cell contacts appears to control the maturation or disassembly of these adhesions and initiates intracellular signaling cascades that ultimately alter many cellular behaviors. In response to externally applied forces, cells actively rearrange the organization and contractile activity of the cytoskeleton and redistribute their intracellular forces. Recent studies suggest that the localized concentration of these cytoskeletal tensions at adhesions is also a major mediator of mechanical signaling. This review summarizes the role of mechanical forces in the formation, stabilization, and dissociation of focal adhesions and adherens junctions and outlines how integration of signals from these adhesions over the entire cell body affects how a cell responds to its mechanical environment. This review also describes advanced optical, lithographic, and computational techniques for the study of mechanotransduction.
Collapse
Affiliation(s)
- Christopher S Chen
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA.
| | | | | |
Collapse
|
200
|
Rajasekaran SA, Gopal J, Espineda C, Ryazantsev S, Schneeberger EE, Rajasekaran AK. HPAF-II, a cell culture model to study pancreatic epithelial cell structure and function. Pancreas 2004; 29:e77-83. [PMID: 15367897 DOI: 10.1097/00006676-200410000-00016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Epithelial cells have distinct apical and basolateral plasma membrane domains separated by tight junctions. This phenotype is essential for the directional transport functions of epithelial cells. Here we characterized a well-differentiated pancreatic epithelial cell line to establish a useful model for understanding the mechanisms involved in the regulation of junctional complexes, polarity, and disease processes in the pancreas. METHODS Immunofluorescence of cell junction marker proteins and electron microscopy were used to determine the presence of tight junctions, adherens junctions, and desmosomes. The functionality of tight junctions was tested by transepithelial resistance measurements and transepithelial permeability studies of nonionic molecules. Tight junction function in polarity was determined by laser scanning confocal microscopy. RESULTS Immunofluorescence analysis in HPAF-II cells revealed tight junction localization of ZO-1, occludin, and claudin-4; adherens junction localization of E-cadherin and beta-catenin; and desmosomal localization of desmocollin. Transmission electron microscopy showed the presence of tight junctions, adherens junctions, and des-mosomes, and freeze-fracture electron microscopy revealed the presence of distinct anastomosing tight junction strands. Transepithelial electrical resistance and permeability measurements revealed functional tight junctions. In addition, 3-dimensional images of the monolayer generated by laser scanning confocal microscopy revealed that HPAF-II cells show polarity. Immunoblotting and RT-PCR analyses revealed high expression levels of E-cadherin and Na,K-ATPase beta-subunit but low levels of the transcription factor Snail in HPAF-II cells compared with MiaPaCa-2 cells. CONCLUSION The HPAF-II cell line is a well-differentiated human pancreatic carcinoma cell line that should be useful as a model for studies aimed at understanding epithelial polarity, regulation of junctional complexes, and disease processes in pancreas.
Collapse
Affiliation(s)
- Sigrid A Rajasekaran
- Department of Pathology and Laboratory Medicine, UCLA David Geffen School of Medicine, Los Angeles, California 90095, USA
| | | | | | | | | | | |
Collapse
|