1
|
Huang J, Luo S, Shen J, Lee M, Chen R, Ma S, Sun LQ, Li JJ. Cellular polarity pilots breast cancer progression and immunosuppression. Oncogene 2025; 44:783-793. [PMID: 40057606 PMCID: PMC11913746 DOI: 10.1038/s41388-025-03324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Disrupted cellular polarity (DCP) is a hallmark of solid cancer, the malignant disease of epithelial tissues, which occupies ~90% of all human cancers. DCP has been identified to affect not only the cancer cell's aggressive behavior but also the migration and infiltration of immune cells, although the precise mechanism of DCP-affected tumor-immune cell interaction remains unclear. This review discusses immunosuppressive tumor microenvironments (TME) caused by DCP-driven tumor cell proliferation with DCP-impaired immune cell functions. We will revisit the fundamental roles of cell polarity (CP) proteins in sustaining mammary luminal homeostasis, epithelial transformation, and breast cancer progression. Then, the current data on CP involvement in immune cell activation, maturation, migration, and tumor infiltration are evaluated. The CP status on the immune effector cells and their targeted tumor cells are highlighted in tumor immune regulation, including the antigen presentation and the formation of immune synapses (IS). CP-regulated antigen presentation and delivery and the formation of IS between the immune cells, especially between the immune effectors and tumor cells, will be addressed. Alterations of CP on the tumor cells, infiltrated immune effector cells, or both are discussed with these aspects. We conclude that CP-mediated tumor aggressiveness coupled with DCP-impaired immune cell disability may decide the degree of immunosuppressive status and responsiveness to immune checkpoint blockade (ICB). Further elucidating the dynamics of CP- or DCP-mediated immune regulation in TME will provide more critical insights into tumor-immune cell dynamics, which is required to invent more effective approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shufeng Luo
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha
| | - Juan Shen
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Maya Lee
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Rachel Chen
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lun-Quan Sun
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha.
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA.
- NCI-designated Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California, USA.
| |
Collapse
|
2
|
Shen Z, Gao Y, Sun X, Chen M, Cen C, Wang M, Wang N, Liu B, Li J, Cui X, Hou J, Shi Y, Gao F. Inactivation of JNK signalling results in polarity loss and cell senescence of Sertoli cell. Cell Prolif 2025; 58:e13760. [PMID: 39329440 PMCID: PMC11839192 DOI: 10.1111/cpr.13760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
As major somatic cells in the testis, Sertoli cell development is precisely regulated by numerous factors, and aberrant development of these cells is associated with male reproductive diseases. JNK signalling is evolutionarily conserved and involved in multiple critical biological processes. Here, we found that the double knockout of Jnk1 and Jnk2 resulted in aberrant localisation of Sertoli cells at early developmental stages, with most Sertoli cells being lost at later stages. Further studies revealed that the inactivation of JNK signalling caused polarity loss in Sertoli cells. In vitro-cultured Jnk1/2-DKO Sertoli cells exhibited a senescence-associated phenotype. Mechanistic studies demonstrate that JNK signalling is likely involved in establishing Sertoli cell polarity by regulating the expression of TGF-β2, mediated by c-Jun. The senescence of Sertoli cells in JNKs-deficient mice is caused by aberrant proteolysis of P27KIP1, mediated by c-Myc. This study demonstrates the role of JNK signalling in Sertoli cell development and functional maintenance, which may also represent an aetiology of male infertility in humans.
Collapse
Affiliation(s)
- Zhiming Shen
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
- Department of Reproductive MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Yang Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xuedong Sun
- Eastern Department of NeurologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Department of NeurologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Changhuo Cen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Mengyue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Nan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Bowen Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jiayi Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiuhong Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jian Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological SciencesChina Agricultural UniversityBeijingChina
| | - Yuhua Shi
- Department of Reproductive MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Department of Obstetrics and Gynecology, Center for Reproductive MedicineNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| |
Collapse
|
3
|
Laumonnerie C, Shamambo M, Stabley DR, Lewis TL, Trivedi N, Howell D, Solecki DJ. Siah2 antagonism of Pard3/JamC modulates Ntn1-Dcc signaling to regulate cerebellar granule neuron germinal zone exit. Nat Commun 2025; 16:355. [PMID: 39774925 PMCID: PMC11706986 DOI: 10.1038/s41467-024-55400-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Exiting a germinal zone (GZ) initiates a cascade of events that promote neuronal maturation and circuit assembly. Developing neurons and their progenitors must interpret various niche signals-such as morphogens, guidance molecules, extracellular matrix components, and adhesive cues-to navigate this region. How differentiating neurons in mouse brains integrate and adapt to multiple cell-extrinsic niche cues with their cell-intrinsic machinery in exiting a GZ is unknown. We establish cooperation between cell polarity-regulated adhesion and Netrin-1 signaling comprises a coincidence detection circuit repelling maturing neurons from their GZ. In this circuit, the Partitioning defective 3 (Pard3) polarity protein and Junctional adhesion molecule-C (JamC) adhesion molecule promote, while the Seven in absentia 2 (Siah2) ubiquitin ligase inhibits, Deleted in colorectal cancer (Dcc) receptor surface recruitment to gate differentiation linked repulsion to GZ Netrin-1. These results demonstrate cell polarity as a central integrator of adhesive- and guidance cues cooperating to spur GZ exit.
Collapse
Affiliation(s)
- Christophe Laumonnerie
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Maleelo Shamambo
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Daniel R Stabley
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Niraj Trivedi
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - Danielle Howell
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA
| | - David J Solecki
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38104, USA.
| |
Collapse
|
4
|
Laumonnerie C, Shamambo M, Stabley DR, Lewis TL, Trivedi N, Howell D, Solecki DJ. Antagonistic action of Siah2 and Pard3/JamC to promote germinal zone exit of differentiated cerebellar granule neurons by modulating Ntn1 signaling via Dcc. RESEARCH SQUARE 2024:rs.3.rs-1819367. [PMID: 39399669 PMCID: PMC11469462 DOI: 10.21203/rs.3.rs-1819367/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Exiting a germinal zone (GZ) initiates a cascade of events that promote neuronal maturation and circuit assembly. Developing neurons and their progenitors must interpret various niche signals-such as morphogens, guidance molecules, extracellular matrix components, and adhesive cues-to navigate this region. How differentiating neurons integrate and adapt to multiple cell-extrinsic niche cues with their cell-intrinsic machinery in exiting a GZ is unknown. We establish cooperation between cell polarity-regulated adhesion and Netrin-1 (Ntn-1) signaling comprises a coincidence detection circuit repelling maturing neurons from their GZ. In this circuit, the Partitioning defective 3 (Pard3) polarity protein and Junctional adhesion molecule-C (JamC) adhesion protein promote, while the Seven in absentia 2 (Siah2) ubiquitin ligase inhibits, Deleted in colorectal cancer (Dcc) receptor surface recruitment to gate differentiation linked repulsion to GZ Ntn-1. These results demonstrate cell polarity as a central integrator of adhesive- and guidance cues cooperating to spur GZ exit.
Collapse
Affiliation(s)
- Christophe Laumonnerie
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Maleelo Shamambo
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Daniel R Stabley
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104
| | - Niraj Trivedi
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - Danielle Howell
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| | - David J Solecki
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38104
| |
Collapse
|
5
|
Wibbe N, Steinbacher T, Tellkamp F, Beckmann N, Brinkmann F, Stecher M, Gerke V, Niessen CM, Ebnet K. RhoGDI1 regulates cell-cell junctions in polarized epithelial cells. Front Cell Dev Biol 2024; 12:1279723. [PMID: 39086660 PMCID: PMC11288927 DOI: 10.3389/fcell.2024.1279723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Cell-cell contact formation of polarized epithelial cells is a multi-step process that involves the co-ordinated activities of Rho family small GTPases. Consistent with the central role of Rho GTPases, a number of Rho guanine nucleotide exchange factors (GEFs) and Rho GTPase-activating proteins (GAPs) have been identified at cell-cell junctions at various stages of junction maturation. As opposed to RhoGEFs and RhoGAPs, the role of Rho GDP dissociation inhibitors (GDIs) during cell-cell contact formation is poorly understood. Here, we have analyzed the role of RhoGDI1/ARHGDIA, a member of the RhoGDI family, during cell-cell contact formation of polarized epithelial cells. Depletion of RhoGDI1 delays the development of linear cell-cell junctions and the formation of barrier-forming tight junctions. In addition, RhoGDI1 depletion impairs the ability of cells to stop migration in response to cell collision and increases the migration velocity of collectively migrating cells. We also find that the cell adhesion receptor JAM-A promotes the recruitment of RhoGDI1 to cell-cell contacts. Our findings implicate RhoGDI1 in various processes involving the dynamic reorganization of cell-cell junctions.
Collapse
Affiliation(s)
- Nicolina Wibbe
- Institute-Associated Research Group “Cell Adhesion and Cell Polarity”, Institute of Medical Biochemistry, Zentrum für Molekularbiologie der Entzündung, University Münster, Münster, Germany
| | - Tim Steinbacher
- Institute-Associated Research Group “Cell Adhesion and Cell Polarity”, Institute of Medical Biochemistry, Zentrum für Molekularbiologie der Entzündung, University Münster, Münster, Germany
| | - Frederik Tellkamp
- Department Cell Biology of the Skin, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Niklas Beckmann
- Institute-Associated Research Group “Cell Adhesion and Cell Polarity”, Institute of Medical Biochemistry, Zentrum für Molekularbiologie der Entzündung, University Münster, Münster, Germany
| | - Frauke Brinkmann
- Institute of Medical Biochemistry, ZMBE, University Münster, Münster, Germany
| | - Manuel Stecher
- Institute of Medical Biochemistry, ZMBE, University Münster, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, ZMBE, University Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, Münster, Germany
| | - Carien M. Niessen
- Department Cell Biology of the Skin, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Department Cell Biology of the Skin, University Hospital of Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group “Cell Adhesion and Cell Polarity”, Institute of Medical Biochemistry, Zentrum für Molekularbiologie der Entzündung, University Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, Münster, Germany
| |
Collapse
|
6
|
Berve K, Michel J, Tietz S, Blatti C, Ivan D, Enzmann G, Lyck R, Deutsch U, Locatelli G, Engelhardt B. Junctional adhesion molecule-A deficient mice are protected from severe experimental autoimmune encephalomyelitis. Eur J Immunol 2024; 54:e2350761. [PMID: 38566526 DOI: 10.1002/eji.202350761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024]
Abstract
In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.
Collapse
Affiliation(s)
- Kristina Berve
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Julia Michel
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Claudia Blatti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Daniela Ivan
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
7
|
Torcq L, Majello S, Vivier C, Schmidt AA. Tuning apicobasal polarity and junctional recycling in the hemogenic endothelium orchestrates the morphodynamic complexity of emerging pre-hematopoietic stem cells. eLife 2024; 12:RP91429. [PMID: 38809590 PMCID: PMC11136496 DOI: 10.7554/elife.91429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Hematopoietic stem cells emerge in the embryo from an aortic-derived tissue called the hemogenic endothelium (HE). The HE appears to give birth to cells of different nature and fate but the molecular principles underlying this complexity are largely unknown. Here we show, in the zebrafish embryo, that two cell types emerge from the aortic floor with radically different morphodynamics. With the support of live imaging, we bring evidence suggesting that the mechanics underlying the two emergence types rely, or not, on apicobasal polarity establishment. While the first type is characterized by reinforcement of apicobasal polarity and maintenance of the apical/luminal membrane until release, the second type emerges via a dynamic process reminiscent of trans-endothelial migration. Interfering with Runx1 function suggests that the balance between the two emergence types depends on tuning apicobasal polarity at the level of the HE. In support of this and unexpectedly, we show that Pard3ba - one of the four Pard3 proteins expressed in the zebrafish - is sensitive to interference with Runx1 activity, in aortic endothelial cells. This supports the idea of a signaling cross talk controlling cell polarity and its associated features, between aortic and hemogenic cells. In addition, using new transgenic fish lines that express Junctional Adhesion Molecules and functional interference, we bring evidence for the essential role of ArhGEF11/PDZ-RhoGEF in controlling the HE-endothelial cell dynamic interface, including cell-cell intercalation, which is ultimately required for emergence completion. Overall, we highlight critical cellular and dynamic events of the endothelial-to-hematopoietic transition that support emergence complexity, with a potential impact on cell fate.
Collapse
Affiliation(s)
- Léa Torcq
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
- Sorbonne UniversitéParisFrance
| | - Sara Majello
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
| | - Catherine Vivier
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
| | - Anne A Schmidt
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris CitéParisFrance
| |
Collapse
|
8
|
Hallada LP, Shirinifard A, Solecki DJ. Junctional Adhesion Molecule (JAM)-C recruitment of Pard3 and drebrin to cell contacts initiates neuron-glia recognition and layer-specific cell sorting in developing cerebella. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586832. [PMID: 38585827 PMCID: PMC10996703 DOI: 10.1101/2024.03.26.586832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Sorting maturing neurons into distinct layers is critical for brain development, with disruptions leading to neurological disorders and pediatric cancers. Lamination coordinates where, when, and how cells interact, facilitating events that direct migrating neurons to their destined positions within emerging neural networks and control the wiring of connections in functional circuits. While the role of adhesion molecule expression and presentation in driving adhesive recognition during neuronal migration along glial fibers is recognized, the mechanisms by which the spatial arrangement of these molecules on the cell surface dictates adhesive specificity and translates contact-based external cues into intracellular responses like polarization and cytoskeletal organization remain largely unexplored. We used the cerebellar granule neuron (CGN) system to demonstrate that JAM-C receptor cis-binding on the same cell and trans-binding to neighboring cells controls the recruitment of the Pard3 polarity protein and drebrin microtubule-actin crosslinker at CGN to glial adhesion sites, complementing previous studies that showed Pard3 controls JAM-C exocytic surface presentation. Leveraging advanced imaging techniques, specific probes for cell recognition, and analytical methods to dissect adhesion dynamics, our findings reveal: 1) JAM-C cis or trans mutants result in reduced adhesion formation between CGNs and cerebellar glia, 2) these mutants exhibit delayed recruitment of Pard3 at the adhesion sites, and 3) CGNs with JAM-C mutations experience postponed sorting and entry into the cerebellar molecular layer (ML). By developing a conditional system to image adhesion components from two different cells simultaneously, we made it possible to investigate the dynamics of cell recognition on both sides of neuron-glial contacts and the subsequent recruitment of proteins required for CGN migration. This system and an approach that calculates local correlation based on convolution kernels at the cell adhesions site revealed that CGN to CGN JAM recognition preferentially recruits higher levels of Pard3 and drebrin than CGN to glia JAM recognition. The long latency time of CGNs in the inner external germinal layer (EGL) can be attributed to the combined strength of CGN-CGN contacts and the less efficient Pard3 recruitment by CGN-BG contacts, acting as gatekeepers to ML entry. As CGNs eventually transition to glia binding for radial migration, our research demonstrates that establishing permissive JAM-recognition sites on glia via cis and trans interactions of CGN JAM-C serves as a critical temporal checkpoint for sorting at the EGL to ML boundary. This mechanism integrates intrinsic and extrinsic cellular signals, facilitating heterotypic cell sorting into the ML and dictating the precise spatial organization within the cerebellar architecture.
Collapse
|
9
|
Liu K, Yang H, Xiong R, Shen Y, Song G, Yang J, Wang Z. Generation and characterization of mAb 61H9 against junctional adhesion molecule-a with potent antitumor activity. PeerJ 2024; 12:e17088. [PMID: 38495763 PMCID: PMC10944630 DOI: 10.7717/peerj.17088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Junctional adhesion molecule-A (JAM-A) is an adhesion molecule that exists on the surface of certain types of cells, including white blood cells, endothelial cells, and dendritic cells. In this study, the cDNA sequences of JAM-A-Fc were chemically synthesized with optimization for mammalian expression. Afterward, we analyzed JAM-A protein expression through transient transfection in HEK293 cell lines. Mice were immunized with JAM-A-Fc protein, and hybridoma was prepared by fusing myeloma cells and mouse spleen cells. Antibodies were purified from the hybridoma supernatant and four monoclonal strains were obtained and numbered 61H9, 70E5, 71A8, and 74H3 via enzyme-linked immunosorbent assay screening. Immunofluorescence staining assay showed 61H9 was the most suitable cell line for mAb production due to its fluorescence signal being the strongest. Flow cytometric analysis proved that 61H9 possessed high affinity. Moreover, antagonism of JAM-A mAb could attenuate the proliferative, migrative, and invasive abilities of ESCC cells and significantly inhibit tumor growth in mice. By examining hematoxylin-eosin staining mice tumor tissues, we found inflammatory cells infiltrated lightly in the anti-JAM-A group. The expression of BCL-2 and IκBα in the anti-JAM-A group were decreased in mice tumor tissues compared to the control group. Ultimately, a method for preparing high-yield JAM-A-Fc protein was created and a high affinity mAb against JAM-A with an antitumor effect was prepared.
Collapse
Affiliation(s)
- Kang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hang Yang
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yunlong Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Guiqin Song
- School of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Zhenling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Mangeol P, Massey-Harroche D, Sebbagh M, Richard F, Le Bivic A, Lenne PF. The zonula adherens matura redefines the apical junction of intestinal epithelia. Proc Natl Acad Sci U S A 2024; 121:e2316722121. [PMID: 38377188 PMCID: PMC10907237 DOI: 10.1073/pnas.2316722121] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Cell-cell apical junctions of epithelia consist of multiprotein complexes that organize as belts regulating cell-cell adhesion, permeability, and mechanical tension: the tight junction (zonula occludens), the zonula adherens (ZA), and the macula adherens. The prevailing dogma is that at the ZA, E-cadherin and catenins are lined with F-actin bundles that support and transmit mechanical tension between cells. Using super-resolution microscopy on human intestinal biopsies and Caco-2 cells, we show that two distinct multiprotein belts are basal of the tight junctions as the intestinal epithelia mature. The most apical is populated with nectins/afadin and lined with F-actin; the second is populated with E-cad/catenins. We name this dual-belt architecture the zonula adherens matura. We find that the apical contraction apparatus and the dual-belt organization rely on afadin expression. Our study provides a revised description of epithelial cell-cell junctions and identifies a module regulating the mechanics of epithelia.
Collapse
Affiliation(s)
- Pierre Mangeol
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM–UMR7288, Marseille13009, France
| | - Dominique Massey-Harroche
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM–UMR7288, Marseille13009, France
| | - Michael Sebbagh
- Aix Marseille Université, INSERM, Dynamics and Nanoenvironment of Biological Membrane, DyNaMo, Turing Center for Living Systems, Marseille 13009, France
| | - Fabrice Richard
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM–UMR7288, Marseille13009, France
| | - André Le Bivic
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM–UMR7288, Marseille13009, France
| | - Pierre-François Lenne
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM–UMR7288, Turing Center for Living Systems, Marseille13009, France
| |
Collapse
|
11
|
Canse C, Yildirim E, Yaba A. Overview of junctional complexes during mammalian early embryonic development. Front Endocrinol (Lausanne) 2023; 14:1150017. [PMID: 37152932 PMCID: PMC10158982 DOI: 10.3389/fendo.2023.1150017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/28/2023] [Indexed: 05/09/2023] Open
Abstract
Cell-cell junctions form strong intercellular connections and mediate communication between blastomeres during preimplantation embryonic development and thus are crucial for cell integrity, polarity, cell fate specification and morphogenesis. Together with cell adhesion molecules and cytoskeletal elements, intercellular junctions orchestrate mechanotransduction, morphokinetics and signaling networks during the development of early embryos. This review focuses on the structure, organization, function and expressional pattern of the cell-cell junction complexes during early embryonic development. Understanding the importance of dynamic junction formation and maturation processes will shed light on the molecular mechanism behind developmental abnormalities of early embryos during the preimplantation period.
Collapse
Affiliation(s)
- Ceren Canse
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Ecem Yildirim
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, Istanbul, Türkiye
| | - Aylin Yaba
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, Istanbul, Türkiye
- *Correspondence: Aylin Yaba,
| |
Collapse
|
12
|
Liang X, Weberling A, Hii CY, Zernicka‐Goetz M, Buckley CE. E-cadherin mediates apical membrane initiation site localisation during de novo polarisation of epithelial cavities. EMBO J 2022; 41:e111021. [PMID: 35993232 PMCID: PMC9753465 DOI: 10.15252/embj.2022111021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 01/15/2023] Open
Abstract
Individual cells within de novo polarising tubes and cavities must integrate their forming apical domains into a centralised apical membrane initiation site (AMIS). This is necessary to enable organised lumen formation within multi-cellular tissue. Despite the well-documented importance of cell division in localising the AMIS, we have found a division-independent mechanism of AMIS localisation that relies instead on Cadherin-mediated cell-cell adhesion. Our study of de novo polarising mouse embryonic stem cells (mESCs) cultured in 3D suggests that cell-cell adhesion localises apical proteins such as PAR-6 to a centralised AMIS. Unexpectedly, we also found that mESC clusters lacking functional E-cadherin still formed a lumen-like cavity in the absence of AMIS localisation but did so at a later stage of development via a "closure" mechanism, instead of via hollowing. This work suggests that there are two, interrelated mechanisms of apical polarity localisation: cell adhesion and cell division. Alignment of these mechanisms in space allows for redundancy in the system and ensures the development of a coherent epithelial structure within a growing organ.
Collapse
Affiliation(s)
- Xuan Liang
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Antonia Weberling
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Chun Yuan Hii
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Magdalena Zernicka‐Goetz
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaCAUSA
| | - Clare E Buckley
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
13
|
Apical-basal polarity and the control of epithelial form and function. Nat Rev Mol Cell Biol 2022; 23:559-577. [PMID: 35440694 DOI: 10.1038/s41580-022-00465-y] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 02/02/2023]
Abstract
Epithelial cells are the most common cell type in all animals, forming the sheets and tubes that compose most organs and tissues. Apical-basal polarity is essential for epithelial cell form and function, as it determines the localization of the adhesion molecules that hold the cells together laterally and the occluding junctions that act as barriers to paracellular diffusion. Polarity must also target the secretion of specific cargoes to the apical, lateral or basal membranes and organize the cytoskeleton and internal architecture of the cell. Apical-basal polarity in many cells is established by conserved polarity factors that define the apical (Crumbs, Stardust/PALS1, aPKC, PAR-6 and CDC42), junctional (PAR-3) and lateral (Scribble, DLG, LGL, Yurt and RhoGAP19D) domains, although recent evidence indicates that not all epithelia polarize by the same mechanism. Research has begun to reveal the dynamic interactions between polarity factors and how they contribute to polarity establishment and maintenance. Elucidating these mechanisms is essential to better understand the roles of apical-basal polarity in morphogenesis and how defects in polarity contribute to diseases such as cancer.
Collapse
|
14
|
Lv T, Xu J, Yuan H, Wang J, Jiang X. Dual Function of Par3 in Tumorigenesis. Front Oncol 2022; 12:915957. [PMID: 35875120 PMCID: PMC9305838 DOI: 10.3389/fonc.2022.915957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022] Open
Abstract
Cell maintenance and the establishment of cell polarity involve complicated interactions among multiple protein complexes as well as the regulation of different signaling pathways. As an important cell polarity protein, Par3 is evolutionarily conserved and involved in tight junction formation as well as tumorigenesis. In this review, we aimed to explore the function of Par3 in tumorigenesis. Research has shown that Par3 exhibits dual functions in human cancers, both tumor-promoting and tumor-suppressive. Here, we focus on the activities of Par3 in different stages and types of tumors, aiming to offer a new perspective on the molecular mechanisms that regulate the functions of Par3 in tumor development. Tumor origin, tumor microenvironment, tumor type, cell density, cell–cell contact, and the synergistic effect of Par3 and other tumor-associated signaling pathways may be important reasons for the dual function of Par3. The important role of Par3 in mammalian tumorigenesis and potential signaling pathways is context dependent.
Collapse
Affiliation(s)
- Tao Lv
- Centre for Yunnan Plateau Biological Resources Protection and Utilization, College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, China
- Yunnan Engineering Research Center of Fruit Wine, Qujing Normal University, Qujing, China
- Key Laboratory of Yunnan Province Universities of Qujing Natural History and Early Vertebrate Evolution, College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, China
| | - Jiashun Xu
- Centre for Yunnan Plateau Biological Resources Protection and Utilization, College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, China
| | - Hemei Yuan
- Centre for Yunnan Plateau Biological Resources Protection and Utilization, College of Biological Resource and Food Engineering, Qujing Normal University, Qujing, China
| | - Jianling Wang
- College of Chemistry and Environmental Science, Qujing Normal University, Qujing, China
- *Correspondence: Jianling Wang, ; Xinni Jiang,
| | - Xinni Jiang
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
- *Correspondence: Jianling Wang, ; Xinni Jiang,
| |
Collapse
|
15
|
Ebnet K, Gerke V. Rho and Rab Family Small GTPases in the Regulation of Membrane Polarity in Epithelial Cells. Front Cell Dev Biol 2022; 10:948013. [PMID: 35859901 PMCID: PMC9289151 DOI: 10.3389/fcell.2022.948013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/14/2022] [Indexed: 11/27/2022] Open
Abstract
Membrane polarity, defined as the asymmetric distribution of lipids and proteins in the plasma membrane, is a critical prerequisite for the development of multicellular tissues, such as epithelia and endothelia. Membrane polarity is regulated by polarized trafficking of membrane components to specific membrane domains and requires the presence of intramembrane diffusion barriers that prevent the intermixing of asymmetrically distributed membrane components. This intramembrane diffusion barrier is localized at the tight junctions (TJs) in these cells. Both the formation of cell-cell junctions and the polarized traffic of membrane proteins and lipids are regulated by Rho and Rab family small GTPases. In this review article, we will summarize the recent developments in the regulation of apico-basal membrane polarity by polarized membrane traffic and the formation of the intramembrane diffusion barrier in epithelial cells with a particular focus on the role of Rho and Rab family small GTPases.
Collapse
Affiliation(s)
- Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
- *Correspondence: Klaus Ebnet, ; Volker Gerke,
| | - Volker Gerke
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
- *Correspondence: Klaus Ebnet, ; Volker Gerke,
| |
Collapse
|
16
|
The Roles of Par3, Par6, and aPKC Polarity Proteins in Normal Neurodevelopment and in Neurodegenerative and Neuropsychiatric Disorders. J Neurosci 2022; 42:4774-4793. [PMID: 35705493 DOI: 10.1523/jneurosci.0059-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 11/21/2022] Open
Abstract
Normal neural circuits and functions depend on proper neuronal differentiation, migration, synaptic plasticity, and maintenance. Abnormalities in these processes underlie various neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Neural development and maintenance are regulated by many proteins. Among them are Par3, Par6 (partitioning defective 3 and 6), and aPKC (atypical protein kinase C) families of evolutionarily conserved polarity proteins. These proteins perform versatile functions by forming tripartite or other combinations of protein complexes, which hereafter are collectively referred to as "Par complexes." In this review, we summarize the major findings on their biophysical and biochemical properties in cell polarization and signaling pathways. We next summarize their expression and localization in the nervous system as well as their versatile functions in various aspects of neurodevelopment, including neuroepithelial polarity, neurogenesis, neuronal migration, neurite differentiation, synaptic plasticity, and memory. These versatile functions rely on the fundamental roles of Par complexes in cell polarity in distinct cellular contexts. We also discuss how cell polarization may correlate with subcellular polarization in neurons. Finally, we review the involvement of Par complexes in neuropsychiatric and neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. While emerging evidence indicates that Par complexes are essential for proper neural development and maintenance, many questions on their in vivo functions have yet to be answered. Thus, Par3, Par6, and aPKC continue to be important research topics to advance neuroscience.
Collapse
|
17
|
Le LTM, Drakulic S, Nyengaard JR, Golas MM, Sander B. Structural Organization of Human Full-Length PAR3 and the aPKC-PAR6 Complex. Mol Biotechnol 2022; 64:1319-1327. [PMID: 35610404 PMCID: PMC9573856 DOI: 10.1007/s12033-022-00504-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
Abstract
The tripartite partition defect (PAR) polarity complex, which includes the proteins PAR3, atypical protein kinase C (aPKC), and PAR6, is a major regulator of cellular polarity. It is highly conserved and expressed in various tissues. Its largest component, PAR3, controls protein–protein interactions of the PAR complex with a variety of interaction partners, and PAR3 self-association is critical for the formation of filament-like structures. However, little is known about the structure of the PAR complex. Here, we purified non-filamentous PAR3 and the aPKC–PAR6 complex and characterized them by single-particle electron microscopy (EM). We expressed and purified an oligomerization-deficient form of PAR3, PAR3V13D,D70K, and the active aPKC–PAR6 dimer. For PAR3, engineering at two positions is sufficient to form stable single particles with a maximum dimension of 20 nm. aPKC–PAR6 forms a complex with a maximum dimension of 13.5 nm that contains single copies of aPKC. Thus, the data present a basis for further high-resolution studies of PAR proteins and PAR complex formation.
Collapse
Affiliation(s)
- Le T M Le
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Srdja Drakulic
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Wilhelm Meyers Allé 3, Building 1233/1234, 8000, Aarhus C, Denmark
| | - Monika M Golas
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Human Genetics, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Bjoern Sander
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Wilhelm Meyers Allé 3, Building 1233/1234, 8000, Aarhus C, Denmark.
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
18
|
Zhao Y, Peng H, Liang L, Li Y, Hu X, Wang B, Xu Y, Chen S. Polarity protein Par3 sensitizes breast cancer to paclitaxel by promoting cell cycle arrest. Breast Cancer Res Treat 2022; 192:75-87. [PMID: 35079981 DOI: 10.1007/s10549-021-06490-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/02/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE Paclitaxel, belongs to tubulin-binding agents (TBAs), shows a great efficacy against breast cancer via stabilizing microtubules. Drug resistance limits its clinical application. Here we aimed to explore a role of Polarity protein Par3 in improving paclitaxel effectiveness. METHODS Breast cancer specimens from 45 patients were collected to study the relationship between Par3 expression and paclitaxel efficacy. The Kaplan-Meier method was used for survival analysis. Cell viability was measured in breast cancer cells (SK-BR-3 and T-47D) with Par3 over-expression or knockdown. The flow cytometry assays were performed to measure cell apoptosis and cell cycle. BrdU incorporation assay and Hoechst 33,258 staining were performed to measure cell proliferation and cell apoptosis, respectively. Immunofluorescence was used to detect microtubule structures. RESULTS Par3 expression was associated with good response of paclitaxel in breast cancer patients. Consistently, Par3 over-expression significantly sensitized breast cancer cells to paclitaxel by promoting cell apoptosis and reducing cell proliferation. In Par3 overexpressing cells upon paclitaxel treatment, we observed intensified cell cycle arrests at metaphase. Further exploration showed that Par3 over-expression stabilized microtubules of breast cancer cells in response to paclitaxel and resists to microtubules instability induced by nocodazole, a microtubule-depolymerizing agent. CONCLUSION Par3 facilitates polymeric forms of tubulin and stabilizes microtubule structure, which aggravates paclitaxel-induced delay at the metaphase-anaphase transition, leading to proliferation inhibition and apoptosis of breast cancer cells. Par3 has a potential role in sensitizing breast cancer cells to paclitaxel, which may provide a more precise assessment of individual treatment and novel therapeutic targets.
Collapse
Affiliation(s)
- Yannan Zhao
- Department of Medical Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.,NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Huitong Peng
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Limiao Liang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Yi Li
- Department of Medical Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.,NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Xichun Hu
- Department of Medical Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China
| | - Biyun Wang
- Department of Medical Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, 270 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
| | - Yingying Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
| | - She Chen
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 130 Dong'an Road, Xuhui District, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
19
|
To Stick or Not to Stick: Adhesions in Orofacial Clefts. BIOLOGY 2022; 11:biology11020153. [PMID: 35205020 PMCID: PMC8869391 DOI: 10.3390/biology11020153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/17/2022]
Abstract
Morphogenesis requires a tight coordination between mechanical forces and biochemical signals to inform individual cellular behavior. For these developmental processes to happen correctly the organism requires precise spatial and temporal coordination of the adhesion, migration, growth, differentiation, and apoptosis of cells originating from the three key embryonic layers, namely the ectoderm, mesoderm, and endoderm. The cytoskeleton and its remodeling are essential to organize and amplify many of the signaling pathways required for proper morphogenesis. In particular, the interaction of the cell junctions with the cytoskeleton functions to amplify the behavior of individual cells into collective events that are critical for development. In this review we summarize the key morphogenic events that occur during the formation of the face and the palate, as well as the protein complexes required for cell-to-cell adhesions. We then integrate the current knowledge into a comprehensive review of how mutations in cell-to-cell adhesion genes lead to abnormal craniofacial development, with a particular focus on cleft lip with or without cleft palate.
Collapse
|
20
|
Huang H, Li Y, Wang L, Song Y, Zhang G. Membrane proteomic analysis identifies the polarity protein PARD3 as a novel antiviral protein against PEDV infection. J Proteomics 2021; 253:104462. [PMID: 34954106 PMCID: PMC8695312 DOI: 10.1016/j.jprot.2021.104462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/01/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a highly pathogenic enteric coronavirus causing lethal watery diarrhea in suckling piglets. PEDV could remodel host membrane structures for their replication, assembly and escape from host cells. However, little is known about the host membrane proteins of PEDV infection. In this study, we analyzed differentially abundant proteins (DAPs) between PEDV infection group and control group and identified the polarity protein PARD3 as one of the most significantly DAPs. PARD3 is implicated in the formation of tight junctions at epithelial cell-cell contacts. Then, we found that PEDV infection promoted the degradation of PARD3 via the ubiquitin proteasome pathway. Moreover, knockdown of PARD3 promoted the proliferation of PEDV. Further study showed that the downregulation of PARD3 altered the normal morphology of the tight junction proteins and promoted apical and basolateral virus proliferation. Tight junctions enable epithelial cells to form physical barriers, which act as an innate immune mechanism that can impede viral infection and PEDV affected the barrier functions by causing degradation of PARD3. Taken together, this work is the first time to investigate the membrane protein profile of PEDV-infected cells using quantitative proteomics and suggests that PARD3 could be a potential novel antiviral protein against PEDV infection. Significance Membrane proteins are involved in various physiological and biochemical functions critical for cellular function. It is also dynamic in nature, where many proteins are changed during in response to environmental stress. However, membrane proteins are difficult to study because of their hydrophobicity. Membrane proteomic methods using mass spectrometry analysis have been developed and applied for the characterization of the plasma membrane and subcellular organelles of various virus infected cells. Porcine epidemic diarrhea virus (PEDV) is an enteric pathogen of importance to the swine industry, causing high mortality in neonatal piglets. Because PEDV infected Vero cells can lead to significant changes in cell membrane morphology and form syncytial lesions. Here, we isolated the membrane proteins of PEDV infected and control cells and applied isobaric tags for relative and absolute quantification (iTRAQ) labeling coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS) to quantitatively identify the differentially abundant proteins (DAPs) in PEDV-infected Vero cells and confirmed the DAPs by performing RT-qPCR and Western blot analysis. Among these differential proteins, we focused on a down-regulated protein PARD3 which is important for cell tight junction and cell polarity. Loss of PARD3 can destroy the tight junction of cells and promote the proliferation of PEDV in the apical and basolateral sides. These findings will provide valuable information to better understand the mechanisms underlying the host defense responses to PEDV infection.
Collapse
Affiliation(s)
- Huimin Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Yongtao Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Li Wang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Huayuan Road No. 116, Zhengzhou 450002, China
| | - Yapeng Song
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Huayuan Road No. 116, Zhengzhou 450002, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
21
|
Marques MS, Costa AC, Osório H, Pinto ML, Relvas S, Dinis-Ribeiro M, Carneiro F, Leite M, Figueiredo C. Helicobacter pylori PqqE is a new virulence factor that cleaves junctional adhesion molecule A and disrupts gastric epithelial integrity. Gut Microbes 2021; 13:1-21. [PMID: 33970782 PMCID: PMC8115454 DOI: 10.1080/19490976.2021.1921928] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Helicobacter pylori infects approximately half of the world's population and is the strongest risk factor for peptic ulcer disease and gastric cancer, representing a major global health concern. H. pylori persistently colonizes the gastric epithelium, where it subverts the highly organized structures that maintain epithelial integrity. Here, a unique strategy used by H. pylori to disrupt the gastric epithelial junctional adhesion molecule-A (JAM-A) is disclosed, using various experimental models that include gastric cell lines, primary human gastric cells, and biopsy specimens of infected and non-infected individuals. H. pylori preferentially cleaves the cytoplasmic domain of JAM-A at Alanine 285. Cells stably transfected with full-length JAM-A or JAM-A lacking the cleaved sequence are used in a range of functional assays, which demonstrate that the H. pylori cleaved region is critical to the maintenance of the epithelial barrier and of cell-cell adhesion. Notably, by combining chromatography techniques and mass spectrometry, PqqE (HP1012) is purified and identified as the H. pylori virulence factor that cleaves JAM-A, uncovering a previously unreported function for this bacterial protease. These findings propose a novel mechanism for H. pylori to disrupt epithelial integrity and functions, breaking new ground in the understanding of the pathogenesis of this highly prevalent and clinically relevant infection.
Collapse
Affiliation(s)
- Miguel S. Marques
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Ana C. Costa
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Hugo Osório
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Marta L. Pinto
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Sandra Relvas
- Department of Pathology, Centro Hospitalar Universitário S. João, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Faculty of Medicine of the University of Porto, Porto, Portugal,Instituto Português de Oncologia, Porto, Portugal,Center for Health Technology and Services Research (CINTESIS), Porto, Portugal
| | - Fátima Carneiro
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal,Department of Pathology, Centro Hospitalar Universitário S. João, Porto, Portugal
| | - Marina Leite
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Ceu Figueiredo
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine of the University of Porto, Porto, Portugal,CONTACT Ceu Figueiredo i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
Vasquez CG, de la Serna EL, Dunn AR. How cells tell up from down and stick together to construct multicellular tissues - interplay between apicobasal polarity and cell-cell adhesion. J Cell Sci 2021; 134:272658. [PMID: 34714332 DOI: 10.1242/jcs.248757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Polarized epithelia define a topological inside and outside, and hence constitute a key evolutionary innovation that enabled the construction of complex multicellular animal life. Over time, this basic function has been elaborated upon to yield the complex architectures of many of the organs that make up the human body. The two processes necessary to yield a polarized epithelium, namely regulated adhesion between cells and the definition of the apicobasal (top-bottom) axis, have likewise undergone extensive evolutionary elaboration, resulting in multiple sophisticated protein complexes that contribute to both functions. Understanding how these components function in combination to yield the basic architecture of a polarized cell-cell junction remains a major challenge. In this Review, we introduce the main components of apicobasal polarity and cell-cell adhesion complexes, and outline what is known about their regulation and assembly in epithelia. In addition, we highlight studies that investigate the interdependence between these two networks. We conclude with an overview of strategies to address the largest and arguably most fundamental unresolved question in the field, namely how a polarized junction arises as the sum of its molecular parts.
Collapse
Affiliation(s)
- Claudia G Vasquez
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Eva L de la Serna
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA.,Biophysics Program, Stanford University, Stanford, CA 94305, USA.,Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
23
|
Czubak-Prowizor K, Babinska A, Swiatkowska M. The F11 Receptor (F11R)/Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A) in cancer progression. Mol Cell Biochem 2021; 477:79-98. [PMID: 34533648 PMCID: PMC8755661 DOI: 10.1007/s11010-021-04259-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022]
Abstract
The F11 Receptor (F11R), also called Junctional Adhesion Molecule-A (JAM-A) (F11R/JAM-A), is a transmembrane glycoprotein of the immunoglobulin superfamily, which is mainly located in epithelial and endothelial cell tight junctions and also expressed on circulating platelets and leukocytes. It participates in the regulation of various biological processes, as diverse as paracellular permeability, tight junction formation and maintenance, leukocyte transendothelial migration, epithelial-to-mesenchymal transition, angiogenesis, reovirus binding, and platelet activation. Dysregulation of F11R/JAM-A may result in pathological consequences and disorders in normal cell function. A growing body of evidence points to its role in carcinogenesis and invasiveness, but its tissue-specific pro- or anti-tumorigenic role remains a debated issue. The following review focuses on the F11R/JAM-A tissue-dependent manner in tumorigenesis and metastasis and also discusses the correlation between poor patient clinical outcomes and its aberrant expression. In the future, it will be required to clarify the signaling pathways that are activated or suppressed via the F11R/JAM-A protein in various cancer types to understand its multiple roles in cancer progression and further use it as a novel direct target for cancer treatment.
Collapse
Affiliation(s)
- Kamila Czubak-Prowizor
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland.
| | - Anna Babinska
- Department of Medicine, State University of New York Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY, 11203, USA
| | - Maria Swiatkowska
- Department of Cytobiology and Proteomics, Medical University of Lodz, 6/8 Mazowiecka St., 92-215, Lodz, Poland
| |
Collapse
|
24
|
Smyth T, Georas SN. Effects of ozone and particulate matter on airway epithelial barrier structure and function: a review of in vitro and in vivo studies. Inhal Toxicol 2021; 33:177-192. [PMID: 34346824 DOI: 10.1080/08958378.2021.1956021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The airway epithelium represents a crucial line of defense against the spread of inhaled pathogens. As the epithelium is the first part of the body to be exposed to the inhaled environment, it must act as both a barrier to and sentinel against any inhaled agents. Despite its vital role in limiting the spread of inhaled pathogens, the airway epithelium is also regularly exposed to air pollutants which disrupt its normal function. Here we review the current understanding of the structure and composition of the airway epithelial barrier, as well as the impact of inhaled pollutants, including the reactive gas ozone and particulate matter, on epithelial function. We discuss the current in vitro, rodent model, and human exposure findings surrounding the impact of various inhaled pollutants on epithelial barrier function, mucus production, and mucociliary clearance. Detailed information on how inhaled pollutants impact epithelial structure and function will further our understanding of the adverse health effects of air pollution exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
25
|
The polarity protein PARD3 and cancer. Oncogene 2021; 40:4245-4262. [PMID: 34099863 DOI: 10.1038/s41388-021-01813-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/10/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Tissue disorganisation is one of the main hallmarks of cancer. Polarity proteins are responsible for the arrangement of cells within epithelial tissues through the asymmetric organisation of cellular components. Partition defective 3 (PARD3) is a master regulator of the Par polarity complex primarily due to its ability to form large complexes via its self-homologous binding domain. In addition to its role in polarity, PARD3 is a scaffolding protein that binds to intracellular signalling molecules, many of which are frequently deregulated in cancer. The role of PARD3 has been implicated in multiple solid cancers as either a tumour suppressor or promoter. This dual functionality is both physiologically and cell context dependent. In this review, we will discuss PARD3's role in tumourigenesis in both laboratory and clinical settings. We will also review several of the mechanisms underpinning PARD3's function including its association with intracellular signalling pathways and its role in the regulation of asymmetric cell division.
Collapse
|
26
|
Development of a Novel Weighted Ranking Method for Immunohistochemical Quantification of a Heterogeneously Expressed Protein in Gastro-Esophageal Cancers. Cancers (Basel) 2021; 13:cancers13061286. [PMID: 33805812 PMCID: PMC7998246 DOI: 10.3390/cancers13061286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 11/29/2022] Open
Abstract
Simple Summary High levels of the protein Junctional Adhesion Molecule-A (JAM-A) have been linked with aggressive disease in patients with several different cancers. However, its distribution is often non-uniform (heterogeneous) across tumors, and can be difficult to quantify. JAM-A has also been linked with high levels of HER2 (an important oncogene) in breast tumors, and the development of resistance to HER2-targeted drugs in those patients. Since gastro-esophageal (GE) cancers are often high in HER2 and have also been approved for HER2-targeted drugs, the aim of this study was to investigate if levels of JAM-A and HER2 are linked in GE cancer. JAM-A was expressed very heterogeneously across miniaturized tissue sections called tissue microarrays (TMAs) of GE cancer patients. In this model, therefore, there was no correlation between JAM-A and HER2 expression. However, when we used larger tissue sections and developed a scoring system to account for heterogeneity, a significant correlation between JAM-A and HER2 levels emerged. This work illustrates the importance of taking intra-tumor heterogeneity into account, particularly in an era when analysis of protein levels by this method is increasingly used to select patients for targeted cancer drugs. Abstract High expression of Junctional Adhesion Molecule-A (JAM-A) has been linked with poor prognosis in several cancers, including breast cancers overexpressing the human epidermal growth factor receptor-2 (HER2). Furthermore, JAM-A expression has been linked with regulating that of HER2, and associated with the development of resistance to HER2-targeted therapies in breast cancer patients. The purpose of this study was to establish a potential relationship between JAM-A and HER2 in HER2-overexpressing gastro-esophageal (GE) cancers. Interrogation of gene expression datasets revealed that high JAM-A mRNA expression was associated with poorer survival in HER2-positive gastric cancer patients. However, high intra-tumoral heterogeneity of JAM-A protein expression was noted upon immunohistochemical scoring of a GE cancer tissue microarray (TMA), precluding a simple confirmation of any relationship between JAM-A and HER2 at protein level. However, in a test-set of 25 full-face GE cancer tissue sections, a novel weighted ranking system proved effective in capturing JAM-A intra-tumoral heterogeneity and confirming statistically significant correlations between JAM-A/HER2 expression. Given the growing importance of immunohistochemistry in stratifying cancer patients for the receipt of new targeted therapies, this may sound a cautionary note against over-relying on cancer TMAs in biomarker discovery studies of heterogeneously expressed proteins. It also highlights a timely need to develop validated mechanisms of capturing intra-tumoral heterogeneity to aid in future biomarker/therapeutic target development for the benefit of cancer patients.
Collapse
|
27
|
MAGIs regulate aPKC to enable balanced distribution of intercellular tension for epithelial sheet homeostasis. Commun Biol 2021; 4:337. [PMID: 33712709 PMCID: PMC7954791 DOI: 10.1038/s42003-021-01874-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 02/19/2021] [Indexed: 01/16/2023] Open
Abstract
Constriction of the apical plasma membrane is a hallmark of epithelial cells that underlies cell shape changes in tissue morphogenesis and maintenance of tissue integrity in homeostasis. Contractile force is exerted by a cortical actomyosin network that is anchored to the plasma membrane by the apical junctional complexes (AJC). In this study, we present evidence that MAGI proteins, structural components of AJC whose function remained unclear, regulate apical constriction of epithelial cells through the Par polarity proteins. We reveal that MAGIs are required to uniformly distribute Partitioning defective-3 (Par-3) at AJC of cells throughout the epithelial monolayer. MAGIs recruit ankyrin-repeat-, SH3-domain- and proline-rich-region-containing protein 2 (ASPP2) to AJC, which modulates Par-3-aPKC to antagonize ROCK-driven contractility. By coupling the adhesion machinery to the polarity proteins to regulate cellular contractility, we propose that MAGIs play essential and central roles in maintaining steady state intercellular tension throughout the epithelial cell sheet. Matsuzawa et al. show that adhesion-related molecules MAGI-1 and MAGI-3 localize partitioning defective-3 (Par-3) at apical junctional complexes of cells throughout the epithelial monolayer. This study provides insights into how tension distribution contributes to cellular contractility in epithelial tissue homeostasis.
Collapse
|
28
|
Martin E, Girardello R, Dittmar G, Ludwig A. New insights into the organization and regulation of the apical polarity network in mammalian epithelial cells. FEBS J 2021; 288:7073-7095. [DOI: 10.1111/febs.15710] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Eleanor Martin
- School of Biological Sciences Nanyang Technological University Singapore City Singapore
- Proteomics of Cellular Signaling Luxembourg Institute of Health Strassen Luxembourg
| | - Rossana Girardello
- School of Biological Sciences Nanyang Technological University Singapore City Singapore
- Proteomics of Cellular Signaling Luxembourg Institute of Health Strassen Luxembourg
| | - Gunnar Dittmar
- Proteomics of Cellular Signaling Luxembourg Institute of Health Strassen Luxembourg
- Department of Life Sciences and Medicine University of Luxembourg Luxembourg
| | - Alexander Ludwig
- School of Biological Sciences Nanyang Technological University Singapore City Singapore
- NTU Institute of Structural Biology (NISB) Experimental Medicine Building Nanyang Technological University Singapore City Singapore
| |
Collapse
|
29
|
Yamashita K, Mizuno K, Furukawa K, Hirose H, Sakurai N, Masuda-Hirata M, Amano Y, Hirose T, Suzuki A, Ohno S. Phosphorylation and dephosphorylation of Ser852 and Ser889 control the clustering, localization and function of PAR3. J Cell Sci 2020; 133:jcs244830. [PMID: 33093242 DOI: 10.1242/jcs.244830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/28/2020] [Indexed: 08/31/2023] Open
Abstract
Cell polarity is essential for various asymmetric cellular events, and the partitioning defective (PAR) protein PAR3 (encoded by PARD3 in mammals) plays a unique role as a cellular landmark to establish polarity. In epithelial cells, PAR3 localizes at the subapical border, such as the tight junction in vertebrates, and functions as an apical determinant. Although we know a great deal about the regulators of PAR3 localization, how PAR3 is concentrated and localized to a specific membrane domain remains an important question to be clarified. In this study, we demonstrate that ASPP2 (also known as TP53BP2), which controls PAR3 localization, links PAR3 and protein phosphatase 1 (PP1). The ASPP2-PP1 complex dephosphorylates a novel phosphorylation site, Ser852, of PAR3. Furthermore, Ser852- or Ser889-unphosphorylatable PAR3 mutants form protein clusters, and ectopically localize to the lateral membrane. Concomitance of clustering and ectopic localization suggests that PAR3 localization is a consequence of local clustering. We also demonstrate that unphosphorylatable forms of PAR3 exhibited a low molecular turnover and failed to coordinate rapid reconstruction of the tight junction, supporting that both the phosphorylated and dephosphorylated states are essential for the functional integrity of PAR3.
Collapse
Affiliation(s)
- Kazunari Yamashita
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Keiko Mizuno
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Kana Furukawa
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Hiroko Hirose
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Natsuki Sakurai
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Maki Masuda-Hirata
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Yoshiko Amano
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Tomonori Hirose
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Atsushi Suzuki
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
- Molecular Cellular Biology Laboratory, Yokohama City University Graduate School of Medical Life Science, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Yokohama City University School of Medicine, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
30
|
Rouaud F, Sluysmans S, Flinois A, Shah J, Vasileva E, Citi S. Scaffolding proteins of vertebrate apical junctions: structure, functions and biophysics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183399. [DOI: 10.1016/j.bbamem.2020.183399] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
|
31
|
Chopyk DM, Grakoui A. Contribution of the Intestinal Microbiome and Gut Barrier to Hepatic Disorders. Gastroenterology 2020; 159:849-863. [PMID: 32569766 PMCID: PMC7502510 DOI: 10.1053/j.gastro.2020.04.077] [Citation(s) in RCA: 282] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/16/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023]
Abstract
Intestinal barrier dysfunction and dysbiosis contribute to development of diseases in liver and other organs. Physical, immunologic, and microbiologic (bacterial, fungal, archaeal, viral, and protozoal) features of the intestine separate its nearly 100 trillion microbes from the rest of the human body. Failure of any aspect of this barrier can result in translocation of microbes into the blood and sustained inflammatory response that promote liver injury, fibrosis, cirrhosis, and oncogenic transformation. Alterations in intestinal microbial populations or their functions can also affect health. We review the mechanisms that regulate intestinal permeability and how changes in the intestinal microbiome contribute to development of acute and chronic liver diseases. We discuss individual components of the intestinal barrier and how these are disrupted during development of different liver diseases. Learning more about these processes will increase our understanding of the interactions among the liver, intestine, and its flora.
Collapse
Affiliation(s)
- Daniel M. Chopyk
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, Georgia; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
32
|
Hartmann C, Schwietzer YA, Otani T, Furuse M, Ebnet K. Physiological functions of junctional adhesion molecules (JAMs) in tight junctions. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183299. [DOI: 10.1016/j.bbamem.2020.183299] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/24/2022]
|
33
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
34
|
Post-translational modifications of tight junction transmembrane proteins and their direct effect on barrier function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183330. [PMID: 32376223 DOI: 10.1016/j.bbamem.2020.183330] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/21/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022]
Abstract
Post-translational modifications (PTMs) such as phosphorylation, ubiquitination or glycosylation are processes affecting the conformation, stability, localization and function of proteins. There is clear evidence that PTMs can act upon tight junction (TJ) proteins, thus modulating epithelial barrier function. Compared to transcriptional or translational regulation, PTMs are rapid and more dynamic processes so in the context of barrier maintenance they might be essential for coping with changing environmental or external impacts. The aim of this review is to extract literature deciphering PTMs in TJ proteins directly contributing to epithelial barrier changes in permeability to ions and macromolecules. It is not intended to cover the entire scope of PTMs in TJ proteins and should rather be understood as a digest of TJ protein modifications directly resulting in the tightening or opening of the epithelial barrier.
Collapse
|
35
|
Krahn MP. Phospholipids of the Plasma Membrane - Regulators or Consequence of Cell Polarity? Front Cell Dev Biol 2020; 8:277. [PMID: 32411703 PMCID: PMC7198698 DOI: 10.3389/fcell.2020.00277] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022] Open
Abstract
Cell polarity is a key feature of many eukaryotic cells, including neurons, epithelia, endothelia and asymmetrically dividing stem cells. Apart from the specific localization of proteins to distinct domains of the plasma membrane, most of these cells exhibit an asymmetric distribution of phospholipids within the plasma membrane too. Notably, research over the last years has revealed that many known conserved regulators of apical-basal polarity in epithelial cells are capable of binding to phospholipids, which in turn regulate the localization and to some extent the function of these proteins. Conversely, phospholipid-modifying enzymes are recruited and controlled by polarity regulators, demonstrating an elaborated balance between asymmetrically localized proteins and phospholipids, which are enriched in certain (micro)domains of the plasma membrane. In this review, we will focus on our current understanding of apical-basal polarity and the implication of phospholipids within the plasma membrane during the cell polarization of epithelia and migrating cells.
Collapse
Affiliation(s)
- Michael P. Krahn
- Department of Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Münster, Germany
| |
Collapse
|
36
|
Marchetti L, Engelhardt B. Immune cell trafficking across the blood-brain barrier in the absence and presence of neuroinflammation. VASCULAR BIOLOGY 2020; 2:H1-H18. [PMID: 32923970 PMCID: PMC7439848 DOI: 10.1530/vb-19-0033] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022]
Abstract
To maintain the homeostatic environment required for proper function of CNS neurons the endothelial cells of CNS microvessels tightly regulate the movement of ions and molecules between the blood and the CNS. The unique properties of these blood vascular endothelial cells are termed blood-brain barrier (BBB) and extend to regulating immune cell trafficking into the immune privileged CNS during health and disease. In general, extravasation of circulating immune cells is a multi-step process regulated by the sequential interaction of adhesion and signalling molecules between the endothelial cells and the immune cells. Accounting for the unique barrier properties of CNS microvessels, immune cell migration across the BBB is distinct and characterized by several adaptations. Here we describe the mechanisms that regulate immune cell trafficking across the BBB during immune surveillance and neuroinflammation, with a focus on the current state-of-the-art in vitro and in vivo imaging observations.
Collapse
Affiliation(s)
- Luca Marchetti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
37
|
Duong CN, Nottebaum AF, Butz S, Volkery S, Zeuschner D, Stehling M, Vestweber D. Interference With ESAM (Endothelial Cell-Selective Adhesion Molecule) Plus Vascular Endothelial-Cadherin Causes Immediate Lethality and Lung-Specific Blood Coagulation. Arterioscler Thromb Vasc Biol 2020; 40:378-393. [DOI: 10.1161/atvbaha.119.313545] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective:
Vascular endothelial (VE)-cadherin is of dominant importance for the formation and stability of endothelial junctions, yet induced gene inactivation enhances vascular permeability in the lung but does not cause junction rupture. This study aims at identifying the junctional adhesion molecule, which is responsible for preventing endothelial junction rupture in the pulmonary vasculature in the absence of VE-cadherin.
Approach and Results:
We have compared the relevance of ESAM (endothelial cell-selective adhesion molecule), JAM (junctional adhesion molecule)-A, PECAM (platelet endothelial cell adhesion molecule)-1, and VE-cadherin for vascular barrier integrity in various mouse tissues. Gene inactivation of ESAM enhanced vascular permeability in the lung but not in the heart, skin, and brain. In contrast, deletion of JAM-A or PECAM-1 did not affect barrier integrity in any of these organs. Blocking VE-cadherin with antibodies caused lethality in ESAM
−/−
mice within 30 minutes but had no such effect in JAM-A
−/−
, PECAM-1
−/−
or wild-type mice. Likewise, induced gene inactivation of VE-cadherin caused rapid lethality only in the absence of ESAM. Ultrastructural analysis revealed that only combined interference with VE-cadherin and ESAM disrupted endothelial junctions and caused massive blood coagulation in the lung. Mechanistically, we could exclude a role of platelet ESAM in coagulation, changes in the expression of other junctional proteins or a contribution of cytoplasmic signaling domains of ESAM.
Conclusions:
Despite well-documented roles of JAM-A and PECAM-1 for the regulation of endothelial junctions, only for ESAM, we detected an essential role for endothelial barrier integrity in a tissue-specific way. In addition, we found that it is ESAM which prevents endothelial junction rupture in the lung when VE-cadherin is absent.
Collapse
Affiliation(s)
- Cao Nguyen Duong
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Astrid F. Nottebaum
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Stefan Butz
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Stefan Volkery
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy and Flow Cytometry Unit (D.Z., M.S.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Stehling
- Electron Microscopy and Flow Cytometry Unit (D.Z., M.S.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
38
|
Heinemann U, Schuetz A. Structural Features of Tight-Junction Proteins. Int J Mol Sci 2019; 20:E6020. [PMID: 31795346 PMCID: PMC6928914 DOI: 10.3390/ijms20236020] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
Tight junctions are complex supramolecular entities composed of integral membrane proteins, membrane-associated and soluble cytoplasmic proteins engaging in an intricate and dynamic system of protein-protein interactions. Three-dimensional structures of several tight-junction proteins or their isolated domains have been determined by X-ray crystallography, nuclear magnetic resonance spectroscopy, and cryo-electron microscopy. These structures provide direct insight into molecular interactions that contribute to the formation, integrity, or function of tight junctions. In addition, the known experimental structures have allowed the modeling of ligand-binding events involving tight-junction proteins. Here, we review the published structures of tight-junction proteins. We show that these proteins are composed of a limited set of structural motifs and highlight common types of interactions between tight-junction proteins and their ligands involving these motifs.
Collapse
Affiliation(s)
- Udo Heinemann
- Macromolecular Structure and Interaction Laboratory, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Anja Schuetz
- Protein Production & Characterization Platform, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
39
|
Díaz-Coránguez M, Liu X, Antonetti DA. Tight Junctions in Cell Proliferation. Int J Mol Sci 2019; 20:E5972. [PMID: 31783547 PMCID: PMC6928848 DOI: 10.3390/ijms20235972] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 12/23/2022] Open
Abstract
Tight junction (TJ) proteins form a continuous intercellular network creating a barrier with selective regulation of water, ion, and solutes across endothelial, epithelial, and glial tissues. TJ proteins include the claudin family that confers barrier properties, members of the MARVEL family that contribute to barrier regulation, and JAM molecules, which regulate junction organization and diapedesis. In addition, the membrane-associated proteins such as MAGUK family members, i.e., zonula occludens, form the scaffold linking the transmembrane proteins to both cell signaling molecules and the cytoskeleton. Most studies of TJ have focused on the contribution to cell-cell adhesion and tissue barrier properties. However, recent studies reveal that, similar to adherens junction proteins, TJ proteins contribute to the control of cell proliferation. In this review, we will summarize and discuss the specific role of TJ proteins in the control of epithelial and endothelial cell proliferation. In some cases, the TJ proteins act as a reservoir of critical cell cycle modulators, by binding and regulating their nuclear access, while in other cases, junctional proteins are located at cellular organelles, regulating transcription and proliferation. Collectively, these studies reveal that TJ proteins contribute to the control of cell proliferation and differentiation required for forming and maintaining a tissue barrier.
Collapse
Affiliation(s)
| | | | - David A. Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI 48105, USA; (M.D.-C.); (X.L.)
| |
Collapse
|
40
|
Castro Dias M, Mapunda JA, Vladymyrov M, Engelhardt B. Structure and Junctional Complexes of Endothelial, Epithelial and Glial Brain Barriers. Int J Mol Sci 2019; 20:E5372. [PMID: 31671721 PMCID: PMC6862204 DOI: 10.3390/ijms20215372] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/25/2019] [Accepted: 10/26/2019] [Indexed: 01/04/2023] Open
Abstract
The homeostasis of the central nervous system (CNS) is ensured by the endothelial, epithelial, mesothelial and glial brain barriers, which strictly control the passage of molecules, solutes and immune cells. While the endothelial blood-brain barrier (BBB) and the epithelial blood-cerebrospinal fluid barrier (BCSFB) have been extensively investigated, less is known about the epithelial and mesothelial arachnoid barrier and the glia limitans. Here, we summarize current knowledge of the cellular composition of the brain barriers with a specific focus on describing the molecular constituents of their junctional complexes. We propose that the brain barriers maintain CNS immune privilege by dividing the CNS into compartments that differ with regard to their role in immune surveillance of the CNS. We close by providing a brief overview on experimental tools allowing for reliable in vivo visualization of the brain barriers and their junctional complexes and thus the respective CNS compartments.
Collapse
Affiliation(s)
| | | | | | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland.
| |
Collapse
|
41
|
Otani T, Nguyen TP, Tokuda S, Sugihara K, Sugawara T, Furuse K, Miura T, Ebnet K, Furuse M. Claudins and JAM-A coordinately regulate tight junction formation and epithelial polarity. J Cell Biol 2019; 218:3372-3396. [PMID: 31467165 PMCID: PMC6781433 DOI: 10.1083/jcb.201812157] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/14/2019] [Accepted: 07/24/2019] [Indexed: 01/03/2023] Open
Abstract
Tight junctions (TJs) establish the epithelial barrier and are thought to form a membrane fence to regulate epithelial polarity, although the roles of TJs in epithelial polarity remain controversial. Claudins constitute TJ strands in conjunction with the cytoplasmic scaffolds ZO-1 and ZO-2 and play pivotal roles in epithelial barrier formation. However, how claudins and other TJ membrane proteins cooperate to organize TJs remains unclear. Here, we systematically knocked out TJ components by genome editing and show that while ZO-1/ZO-2-deficient cells lacked TJ structures and epithelial barriers, claudin-deficient cells lacked TJ strands and an electrolyte permeability barrier but formed membrane appositions and a macromolecule permeability barrier. Moreover, epithelial polarity was disorganized in ZO-1/ZO-2-deficient cells, but not in claudin-deficient cells. Simultaneous deletion of claudins and a TJ membrane protein JAM-A resulted in a loss of membrane appositions and a macromolecule permeability barrier and in sporadic epithelial polarity defects. These results demonstrate that claudins and JAM-A coordinately regulate TJ formation and epithelial polarity.
Collapse
Affiliation(s)
- Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Thanh Phuong Nguyen
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Shinsaku Tokuda
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Kei Sugihara
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taichi Sugawara
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Kyoko Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Takashi Miura
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity," Institute of Medical Biochemistry, Zentrum für Molekularbiologie der Entzündung, University of Münster, Münster, Germany
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
42
|
Mundi S, Massaro M, Scoditti E, Carluccio MA, van Hinsbergh VWM, Iruela-Arispe ML, De Caterina R. Endothelial permeability, LDL deposition, and cardiovascular risk factors-a review. Cardiovasc Res 2019; 114:35-52. [PMID: 29228169 DOI: 10.1093/cvr/cvx226] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022] Open
Abstract
Early atherosclerosis features functional and structural changes in the endothelial barrier function that affect the traffic of molecules and solutes between the vessel lumen and the vascular wall. Such changes are mechanistically related to the development of atherosclerosis. Proatherogenic stimuli and cardiovascular risk factors, such as dyslipidaemias, diabetes, obesity, and smoking, all increase endothelial permeability sharing a common signalling denominator: an imbalance in the production/disposal of reactive oxygen species (ROS), broadly termed oxidative stress. Mostly as a consequence of the activation of enzymatic systems leading to ROS overproduction, proatherogenic factors lead to a pro-inflammatory status that translates in changes in gene expression and functional rearrangements, including changes in the transendothelial transport of molecules, leading to the deposition of low-density lipoproteins (LDL) and the subsequent infiltration of circulating leucocytes in the intima. In this review, we focus on such early changes in atherogenesis and on the concept that proatherogenic stimuli and risk factors for cardiovascular disease, by altering the endothelial barrier properties, co-ordinately trigger the accumulation of LDL in the intima and ultimately plaque formation.
Collapse
Affiliation(s)
- Santa Mundi
- Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, via Monteroni, 73100, Lecce, Italy
| | - Marika Massaro
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Maria Annunziata Carluccio
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Victor W M van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat, NL-1081 BT, Amsterdam, The Netherlands
| | - Marial Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California, 610 Charles E Young Dr S, 90095, Los Angeles, USA; and
| | - Raffaele De Caterina
- Department of Neuroscience, Imaging and Clinical Science and Institute of Advanced Biomedical Technologies, University G. D'Annunzio, via dei Vestini, 66100 Chieti, Italy
| |
Collapse
|
43
|
Shi J, Barakat M, Chen D, Chen L. Bicellular Tight Junctions and Wound Healing. Int J Mol Sci 2018; 19:ijms19123862. [PMID: 30518037 PMCID: PMC6321209 DOI: 10.3390/ijms19123862] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022] Open
Abstract
Bicellular tight junctions (TJs) are intercellular junctions comprised of a variety of transmembrane proteins including occludin, claudins, and junctional adhesion molecules (JAMs) as well as intracellular scaffold proteins such as zonula occludens (ZOs). TJs are functional, intercellular structures that form a barrier between adjacent cells, which constantly seals and unseals to control the paracellular passage of molecules. They are primarily present in the epithelial and endothelial cells of all tissues and organs. In addition to their well-recognized roles in maintaining cell polarity and barrier functions, TJs are important regulators of signal transduction, which modulates cell proliferation, migration, and differentiation, as well as some components of the immune response and homeostasis. A vast breadth of research data is available on TJs, but little has been done to decipher their specific roles in wound healing, despite their primary distribution in epithelial and endothelial cells, which are essential contributors to the wound healing process. Some data exists to indicate that a better understanding of the functions and significance of TJs in healing wounds may prove crucial for future improvements in wound healing research and therapy. Specifically, recent studies demonstrate that occludin and claudin-1, which are two TJ component proteins, are present in migrating epithelial cells at the wound edge but are absent in chronic wounds. This indicates that functional TJs may be critical for effective wound healing. A tremendous amount of work is needed to investigate their roles in barrier function, re-epithelialization, angiogenesis, scar formation, and in the interactions between epithelial cells, endothelial cells, and immune cells both in the acute wound healing process and in non-healing wounds. A more thorough understanding of TJs in wound healing may shed new light on potential research targets and reveal novel strategies to enhance tissue regeneration and improve wound repair.
Collapse
Affiliation(s)
- Junhe Shi
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - Dandan Chen
- Colgate-Palmolive Company, Piscataway, NJ 08855, USA.
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| |
Collapse
|
44
|
Tietz S, Périnat T, Greene G, Enzmann G, Deutsch U, Adams R, Imhof B, Aurrand-Lions M, Engelhardt B. Lack of junctional adhesion molecule (JAM)-B ameliorates experimental autoimmune encephalomyelitis. Brain Behav Immun 2018; 73:3-20. [PMID: 29920328 DOI: 10.1016/j.bbi.2018.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/11/2018] [Accepted: 06/15/2018] [Indexed: 12/28/2022] Open
Abstract
In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) autoaggressive CD4+ T cells cross the blood-brain barrier (BBB) and cause neuroinflammation. Therapeutic targeting of CD4+ T-cell trafficking into the CNS by blocking α4-integrins has proven beneficial for the treatment of MS but comes with associated risks, probably due to blocking CD8+ T cell mediated CNS immune surveillance. Our recent observations show that CD8+ T cells also rely on α4β1-integrins to cross the BBB. Besides vascular cell adhesion molecule-1 (VCAM-1), we identified junctional adhesion molecule-B (JAM-B) as a novel vascular α4β1-integrin ligand involved in CD8+ T-cell migration across the BBB. This prompted us to investigate, if JAM-B also mediates CD4+ T-cell migration across the BBB. We first ensured that encephalitogenic T cells can bind to JAM-B in vitro and next compared EAE pathogenesis in JAM-B-/- C57BL/6J mice and their wild-type littermates. Following immunization with MOGaa35-55 peptide, JAM-B-/- mice developed ameliorated EAE compared to their wild-type littermates. At the same time, we isolated higher numbers of CD45+ infiltrating immune cells from the CNS of JAM-B-/- C57BL/6J mice suffering from EAE. Immunofluorescence staining revealed that the majority of CD45+ inflammatory cells accumulated in the leptomeningeal and perivascular spaces of the CNS behind the BBB but do not gain access to the CNS parenchyma. Trapping of CNS inflammatory cells was not due to increased inflammatory cell proliferation. Neither a loss of BBB integrity or BBB polarity potentially affecting local chemokine gradients nor a lack of focal gelatinase activation required for CNS parenchymal immune cell entry across the glia limitans could be detected in JAM-B-/- mice. Lack of a role for JAM-B in the effector phase of EAE was supported by the observation that we did not detect any role for JAM-B in EAE pathogenesis, when EAE was elicited by in vitro activated MOG aa35-55-specific CD4+ effector T cells. On the other hand, we also failed to demonstrate any role of JAM-B in in vivo priming, proliferation or polarization of MOGaa35-55-specific CD4+ T cells in peripheral immune organs. Finally, our study excludes expression of and thus a role for JAM-B on peripheral and CNS infiltrating myeloid cells. Taken together, although endothelial JAM-B is not required for immune cell trafficking across the BBB in EAE, in its absence accumulation of inflammatory cells mainly in CNS leptomeningeal spaces leads to amelioration of EAE.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Cell Movement/physiology
- Central Nervous System/metabolism
- Central Nervous System/physiology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Endothelium, Vascular/metabolism
- Female
- Integrin alpha4beta1/metabolism
- Junctional Adhesion Molecule B/genetics
- Junctional Adhesion Molecule B/metabolism
- Junctional Adhesion Molecule B/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/physiopathology
- Myelin-Oligodendrocyte Glycoprotein/pharmacology
- Myeloid Cells/metabolism
- Myeloid Cells/physiology
- Tight Junctions/metabolism
Collapse
Affiliation(s)
- Silvia Tietz
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Therese Périnat
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gretchen Greene
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ralf Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Beat Imhof
- Department of Pathology and Immunology, University of Geneva, CMU Geneva, Switzerland
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancerologie de Marseille, INSERM, CNRS, Aix-Marseille University, Marseille, France
| | | |
Collapse
|
45
|
Chen J, Sayadian AC, Lowe N, Lovegrove HE, St Johnston D. An alternative mode of epithelial polarity in the Drosophila midgut. PLoS Biol 2018; 16:e3000041. [PMID: 30339698 PMCID: PMC6209374 DOI: 10.1371/journal.pbio.3000041] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/31/2018] [Accepted: 10/02/2018] [Indexed: 11/18/2022] Open
Abstract
Apical-basal polarity is essential for the formation and function of epithelial tissues, whereas loss of polarity is a hallmark of tumours. Studies in Drosophila have identified conserved polarity factors that define the apical (Crumbs, Stardust, Par-6, atypical protein kinase C [aPKC]), junctional (Bazooka [Baz]/Par-3), and basolateral (Scribbled [Scrib], Discs large [Dlg], Lethal [2] giant larvae [Lgl]) domains of epithelial cells. Because these conserved factors mark equivalent domains in diverse types of vertebrate and invertebrate epithelia, it is generally assumed that this system underlies polarity in all epithelia. Here, we show that this is not the case, as none of these canonical factors are required for the polarisation of the endodermal epithelium of the Drosophila adult midgut. Furthermore, like vertebrate epithelia but not other Drosophila epithelia, the midgut epithelium forms occluding junctions above adherens junctions (AJs) and requires the integrin adhesion complex for polarity. Thus, Drosophila contains two types of epithelia that polarise by fundamentally different mechanisms. This diversity of epithelial types may reflect their different developmental origins, junctional arrangement, or whether they polarise in an apical-basal direction or vice versa. Since knock-outs of canonical polarity factors in vertebrates often have little or no effect on epithelial polarity and the Drosophila midgut shares several common features with vertebrate epithelia, this diversity of polarity mechanisms is likely to be conserved in other animals.
Collapse
Affiliation(s)
- Jia Chen
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Aram-Christopher Sayadian
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Nick Lowe
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Holly E. Lovegrove
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Daniel St Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
46
|
Chuykin I, Ossipova O, Sokol SY. Par3 interacts with Prickle3 to generate apical PCP complexes in the vertebrate neural plate. eLife 2018; 7:37881. [PMID: 30256191 PMCID: PMC6175575 DOI: 10.7554/elife.37881] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/25/2018] [Indexed: 01/15/2023] Open
Abstract
Vertebrate neural tube formation depends on the coordinated orientation of cells in the tissue known as planar cell polarity (PCP). In the Xenopus neural plate, PCP is marked by the enrichment of the conserved proteins Prickle3 and Vangl2 at anterior cell boundaries. Here we show that the apical determinant Par3 is also planar polarized in the neuroepithelium, suggesting a role for Par3 in PCP. Consistent with this hypothesis, interference with Par3 activity inhibited asymmetric distribution of PCP junctional complexes and caused neural tube defects. Importantly, Par3 physically associated with Prickle3 and promoted its apical localization, whereas overexpression of a Prickle3-binding Par3 fragment disrupted PCP in the neural plate. We also adapted proximity biotinylation assay for use in Xenopus embryos and show that Par3 functions by enhancing the formation of the anterior apical PCP complex. These findings describe a mechanistic link between the apical localization of PCP components and morphogenetic movements underlying neurulation.
Collapse
Affiliation(s)
- Ilya Chuykin
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Olga Ossipova
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
47
|
Szymborska A, Gerhardt H. Hold Me, but Not Too Tight-Endothelial Cell-Cell Junctions in Angiogenesis. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029223. [PMID: 28851748 DOI: 10.1101/cshperspect.a029223] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial cell-cell junctions must perform seemingly incompatible tasks during vascular development-providing stable connections that prevent leakage, while allowing dynamic cellular rearrangements during sprouting, anastomosis, lumen formation, and functional remodeling of the vascular network. This review aims to highlight recent insights into the molecular mechanisms governing endothelial cell-cell adhesion in the context of vascular development.
Collapse
Affiliation(s)
- Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,Vascular Patterning Laboratory, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, 3000 Leuven, Belgium.,DZHK (German Centre for Cardiovascular Research), partner site Berlin.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| |
Collapse
|
48
|
Steinbacher T, Kummer D, Ebnet K. Junctional adhesion molecule-A: functional diversity through molecular promiscuity. Cell Mol Life Sci 2018; 75:1393-1409. [PMID: 29238845 PMCID: PMC11105642 DOI: 10.1007/s00018-017-2729-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 12/27/2022]
Abstract
Cell adhesion molecules (CAMs) of the immunoglobulin superfamily (IgSF) regulate important processes such as cell proliferation, differentiation and morphogenesis. This activity is primarily due to their ability to initiate intracellular signaling cascades at cell-cell contact sites. Junctional adhesion molecule-A (JAM-A) is an IgSF-CAM with a short cytoplasmic tail that has no catalytic activity. Nevertheless, JAM-A is involved in a variety of biological processes. The functional diversity of JAM-A resides to a large part in a C-terminal PDZ domain binding motif which directly interacts with nine different PDZ domain-containing proteins. The molecular promiscuity of its PDZ domain motif allows JAM-A to recruit protein scaffolds to specific sites of cell-cell adhesion and to assemble signaling complexes at those sites. Here, we review the molecular characteristics of JAM-A, including its dimerization, its interaction with scaffolding proteins, and the phosphorylation of its cytoplasmic domain, and we describe how these characteristics translate into diverse biological activities.
Collapse
Affiliation(s)
- Tim Steinbacher
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group: Cell Adhesion and Cell Polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Von-Esmarch-Str. 56, 48149, Münster, Germany.
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany.
- Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany.
| |
Collapse
|
49
|
Renschler FA, Bruekner SR, Salomon PL, Mukherjee A, Kullmann L, Schütz-Stoffregen MC, Henzler C, Pawson T, Krahn MP, Wiesner S. Structural basis for the interaction between the cell polarity proteins Par3 and Par6. Sci Signal 2018; 11:11/517/eaam9899. [PMID: 29440511 DOI: 10.1126/scisignal.aam9899] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Polarity is a fundamental property of most cell types. The Par protein complex is a major driving force in generating asymmetrically localized protein networks and consists of atypical protein kinase C (aPKC), Par3, and Par6. Dysfunction of this complex causes developmental abnormalities and diseases such as cancer. We identified a PDZ domain-binding motif in Par6 that was essential for its interaction with Par3 in vitro and for Par3-mediated membrane localization of Par6 in cultured cells. In fly embryos, we observed that the PDZ domain-binding motif was functionally redundant with the PDZ domain in targeting Par6 to the cortex of epithelial cells. Our structural analyses by x-ray crystallography and NMR spectroscopy showed that both the PDZ1 and PDZ3 domains but not the PDZ2 domain in Par3 engaged in a canonical interaction with the PDZ domain-binding motif in Par6. Par3 thus has the potential to recruit two Par6 proteins simultaneously, which may facilitate the assembly of polarity protein networks through multivalent PDZ domain interactions.
Collapse
Affiliation(s)
- Fabian A Renschler
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Susanne R Bruekner
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Paulin L Salomon
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Amrita Mukherjee
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Lars Kullmann
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | | | - Christine Henzler
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Tony Pawson
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Michael P Krahn
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany.,Medical Clinic D, University Hospital of Münster, Domagkstraβe 3a, 48149 Münster, Germany
| | - Silke Wiesner
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany.
| |
Collapse
|
50
|
Kakuki T, Kurose M, Takano KI, Kondoh A, Obata K, Nomura K, Miyata R, Kaneko Y, Konno T, Takahashi S, Hatakeyama T, Kohno T, Himi T, Kojima T. Dysregulation of junctional adhesion molecule-A via p63/GATA-3 in head and neck squamous cell carcinoma. Oncotarget 2017; 7:33887-900. [PMID: 27036044 PMCID: PMC5085126 DOI: 10.18632/oncotarget.8432] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 03/07/2016] [Indexed: 11/25/2022] Open
Abstract
Junctional adhesion molecule-A (JAM-A), which belongs to the IgG superfamily, is a tight junction molecule associated with epithelial and endothelial barrier function. Overexpression of JAM-A is also closely associated with invasion and metastasis of cancers such as breast cancer, lung cancer and pancreatic cancer. However, little is known about the mechanism in overexpression of JAM-A in head and neck squamous cell carcinoma (HNSCC). In the present study, we found high expression of JAM-A at the protein and mRNA levels in HNSCC tissues, including those of the oropharynx, larynx, and hypopharynx, together with high protein expression of β-catenin, p63, ΔNp63 and GATA-3. Furthermore, in ELISA, a significant increase of soluble JAM-A in the sera of HNSCC patients was observed compared to healthy subjects. Knockdown of JAM-A by siRNA inhibited cell proliferation, invasion and migration in the HNSCC cell line Detroit562 in vitro. JAM-A expression in Detroit562 was increased via a distinct signal transduction pathway including NF-κB. Expression of JAM-A, β-catenin, p63 and ΔNp63 in Detroit562 was decreased under hypoxia. Knockdown of p63, ΔNp63 or GATA-3 by siRNAs reduced JAM-A expression in Detroit562. In primary cultured HNSCC cells in which CK7, p63, ΔNp63 and GATA-3 were detected, JAM-A expression was decreased by knockdown of p63 or ΔNp63. These results indicate that JAM-A is a biomarker of malignancy in HNSCC and that plasma soluble JAM-A may contribute to serum-based diagnosis of HNSCC. The mechanism of dysregulation of JAM-A via p63/GATA-3 is important in possible molecular targeted therapy for HNSCC.
Collapse
Affiliation(s)
- Takuya Kakuki
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Makoto Kurose
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ken-Ichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Atsushi Kondoh
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kazufumi Obata
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kazuaki Nomura
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ryo Miyata
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yakuto Kaneko
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Syunta Takahashi
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Tsubasa Hatakeyama
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| |
Collapse
|