1
|
Elsaid KA, Zhang LX, Zhao T, Marks A, Jenkins D, Schmidt TA, Jay GD. Proteoglycan 4 (Lubricin) and Regulation of Xanthine Oxidase in Synovial Macrophage as A Mechanism of Controlling Synovitis. RESEARCH SQUARE 2024:rs.3.rs-4934175. [PMID: 39372933 PMCID: PMC11451733 DOI: 10.21203/rs.3.rs-4934175/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: 10/08/2024]
Abstract
Background Synovial macrophages (SMs) are important effectors of joint health and disease. A novel Cx3CR1 + TREM2 + SM population expressing the tight junction protein claudin-5, was recently discovered in synovial lining. Ablation of these SMs was associated with onset of arthritis. Proteoglycan 4 (PRG4) is a mucinous glycoprotein that fulfills lubricating and homeostatic roles in the joint. The aim of this work is to study the role of PRG4 in modulating synovitis in the context of SM homeostasis and assess the contribution of xanthine oxidase (XO)-hypoxia inducible factor alpha (HIF-1α) axis to this regulation. Methods We used Prg4 FrlioxP/FrtloxP ;R26 FlPoER/+ , a novel transgenic mouse, where the Prg4 Frt allele normally expresses the PRG4 protein and was designed to flank the first two exons of Prg4 with a flippase recognition target and "LOXP" sites. Inducing flippase activity with tamoxifen (TAM) inactivates the Frt allele and thus creates a conditional knockout state. We studied anti-inflammatory SMs and XO by quantitative immunohistochemistry, isolated RNA and studied immune pathway activations by multiplexed assays and isolated SMs and studied PRG4 signaling dysfunction in relation to glycolytic switching due to pro-inflammatory activation. Prg4 inactivated mice were treated with oral febuxostat, a specific XO inhibitor, and quantification of Cx3CR1 + TREM2 + SMs, XO immunostaining and synovitis assessment were conducted. Results Prg4 inactivation induced Cx3CR1 + TREM2 + SM loss (p < 0.001) and upregulated glycolysis and innate immune pathways in the synovium. In isolated SMs, Xdh (p < 0.01) and Hif1a (p < 0.05) were upregulated. Pro-inflammatory activation of SMs was evident by enhanced glycolytic flux and XO-generated reactive oxygen species (ROS). Febuxostat reduced glycolytic flux (p < 0.001) and HIF-1α levels (p < 0.0001) in SMs. Febuxostat also reduced systemic inflammation (p < 0.001), synovial hyperplasia (p < 0.001) and preserved Cx3CR1 + TREM2 + SMs (p < 0.0001) in synovia of Prg4 inactivated mice. Conclusions PRG4 is a biologically significant modulator of synovial homeostasis via inhibition of XO expression and downstream HIF-1a activation. PRG4 signaling is anti-inflammatory and promotes synovial homeostasis in chronic synovitis, where direct XO inhibition is potentially therapeutic in chronic synovitis.
Collapse
|
2
|
Babula A, Gałuszka-Bulaga A, Węglarczyk K, Siedlar M, Baj-Krzyworzeka M. CD44‑hyaluronan axis plays a role in the interactions between colon cancer‑derived extracellular vesicles and human monocytes. Oncol Lett 2023; 26:413. [PMID: 37600336 PMCID: PMC10436155 DOI: 10.3892/ol.2023.13999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
During tumor progression, monocytes circulating in the blood or infiltrating tissue may be exposed to tumor-derived extracellular vesicles (TEVs). The first stage of such interactions involves binding of TEVs to the surface of monocytes, followed by their internalization. The present study examines the role of CD44 molecules in the interactions between monocytes and EVs derived from colon cancer cell lines (HCT116 and SW1116). The efficiency of the attachment and engulfment of TEVs by monocytes is linked to the number of TEVs and time of exposure/interaction. The two investigated TEVs, TEVsHCT116 and TEVsSW1116, originating from HCT116 and SW1116 cells, respectively, differ in hyaluronan (HA) cargo, which reflects HA secretion by parental cancer cells. HA-rich TEVsHCT116 are internalized more effectively in comparison with HA-low TEVsSW1116. Blocking of CD44 molecules on monocytes by anti-CD44 monoclonal antibody significantly decreased the engulfment of TEVsHCT116 but not that of TEVsSW1116 after 30 min contact, suggesting the involvement of the HA-CD44 axis. The three subsets of monocytes, classical, intermediate and non-classical, characterized by gradual changes in the expression of CD14 and CD16 markers, also differ in the expression of CD44. The highest expression of CD44 molecules was observed in the intermediate monocyte subset. Blocking of CD44 molecules decreased the internalization of HA-rich TEVs in all three subsets, which is associated with CD44 expression level. It was hypothesized that HA carried by TEVs, potentially as a component of the 'corona' coating, may facilitate the interaction between subsets of monocytes and TEVs, which may influence the fate of TEVs (such as the rate of TEVs adhesion and engulfment) and change monocyte activity.
Collapse
Affiliation(s)
- Aneta Babula
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 31-530 Kraków, Poland
| | - Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland
| |
Collapse
|
3
|
Weng X, Maxwell-Warburton S, Hasib A, Ma L, Kang L. The membrane receptor CD44: novel insights into metabolism. Trends Endocrinol Metab 2022; 33:318-332. [PMID: 35249813 DOI: 10.1016/j.tem.2022.02.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
Abstract
CD44, a cell-surface glycoprotein, has long been studied as a cancer molecule due to its essential role in physiological activities in normal cells and pathological activities in cancer cells, such as cell proliferation, adhesion, and migration; angiogenesis; inflammation; and cytoskeleton rearrangement. Yet, recent evidence suggests a role of CD44 in metabolism, especially insulin resistance in obesity and diabetes. In line with the current concept of fibroinflammation in obesity and insulin resistance, CD44 as the main receptor of the extracellular matrix component, hyaluronan (HA), has been shown to regulate diet-induced insulin resistance in muscle and other insulin-sensitive tissues. In this review, we integrate current evidence for a role of CD44 in regulating glucose and lipid homeostasis and speculate about its involvement in the pathogenesis of chronic metabolic diseases, including obesity and diabetes. We summarize the current development of CD44-targeted therapies and discuss its potential for the use in treating metabolic diseases.
Collapse
Affiliation(s)
- Xiong Weng
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | | | - Annie Hasib
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Lifeng Ma
- School of Medicine, Xizang Minzhu University, Xianyang, Shaanxi, China
| | - Li Kang
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK.
| |
Collapse
|
4
|
Linsenmeier L, Mohammadi B, Shafiq M, Frontzek K, Bär J, Shrivastava AN, Damme M, Song F, Schwarz A, Da Vela S, Massignan T, Jung S, Correia A, Schmitz M, Puig B, Hornemann S, Zerr I, Tatzelt J, Biasini E, Saftig P, Schweizer M, Svergun D, Amin L, Mazzola F, Varani L, Thapa S, Gilch S, Schätzl H, Harris DA, Triller A, Mikhaylova M, Aguzzi A, Altmeppen HC, Glatzel M. Ligands binding to the prion protein induce its proteolytic release with therapeutic potential in neurodegenerative proteinopathies. SCIENCE ADVANCES 2021; 7:eabj1826. [PMID: 34818048 PMCID: PMC8612689 DOI: 10.1126/sciadv.abj1826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 05/07/2023]
Abstract
The prion protein (PrPC) is a central player in neurodegenerative diseases, such as prion diseases or Alzheimer’s disease. In contrast to disease-promoting cell surface PrPC, extracellular fragments act neuroprotective by blocking neurotoxic disease-associated protein conformers. Fittingly, PrPC release by the metalloprotease ADAM10 represents a protective mechanism. We used biochemical, cell biological, morphological, and structural methods to investigate mechanisms stimulating this proteolytic shedding. Shed PrP negatively correlates with prion conversion and is markedly redistributed in murine brain in the presence of prion deposits or amyloid plaques, indicating a sequestrating activity. PrP-directed ligands cause structural changes in PrPC and increased shedding in cells and organotypic brain slice cultures. As an exception, some PrP-directed antibodies targeting repetitive epitopes do not cause shedding but surface clustering, endocytosis, and degradation of PrPC. Both mechanisms may contribute to beneficial actions described for PrP-directed ligands and pave the way for new therapeutic strategies against currently incurable neurodegenerative diseases.
Collapse
Affiliation(s)
- Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Julia Bär
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Amulya N. Shrivastava
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM, CNRS, PSL Research University, Paris, France
| | - Markus Damme
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Alexander Schwarz
- Institute of Nanostructure and Solid State Physics, Universität Hamburg, Hamburg, Germany
| | - Stefano Da Vela
- European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Tania Massignan
- Dulbecco Telethon Laboratory of Prions and Amyloids, CIBIO, University of Trento, Trento, Italy
| | - Sebastian Jung
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Angela Correia
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation, UKE, Hamburg, Germany
| | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jörg Tatzelt
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Bochum, Germany
| | - Emiliano Biasini
- Dulbecco Telethon Laboratory of Prions and Amyloids, CIBIO, University of Trento, Trento, Italy
| | - Paul Saftig
- Institute of Biochemistry, Christian Albrechts University, Kiel, Germany
| | | | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Ladan Amin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Federica Mazzola
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Simrika Thapa
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - Sabine Gilch
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - Hermann Schätzl
- Calgary Prion Research Unit, University of Calgary, Calgary, Alberta, Canada
| | - David A. Harris
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM, CNRS, PSL Research University, Paris, France
| | - Marina Mikhaylova
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Hermann C. Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
5
|
CD44 and Tumor-Derived Extracellular Vesicles (TEVs). Possible Gateway to Cancer Metastasis. Int J Mol Sci 2021; 22:ijms22031463. [PMID: 33540535 PMCID: PMC7867195 DOI: 10.3390/ijms22031463] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer metastasis, the final stage of tumor progression, is a complex process governed by the interplay of multiple types of cells and the tumor microenvironment. One of the aspects of this interplay involves the release of various factors by the tumor cells alone or by forcing other cells to do so. As a consequence of these actions, tumor cells are prepared in favorable conditions for their dissemination and spread to other sites/organs, which guarantees their escape from immunosurveillance and further progression. Tumor-derived extracellular vesicles (TEVs) represent a heterogeneous population of membrane-bound vesicles that are being actively released by different tumors. The array of proteins (i.e., receptors, cytokines, chemokines, etc.) and nucleic acids (i.e., mRNA, miR, etc.) that TEVs can transfer to other cells is often considered beneficial for the tumor’s survival and proliferation. One of the proteins that is associated with many different tumors as well as their TEVs is a cluster of differentiation 44 in its standard (CD44s) and variant (CD44v) form. This review covers the present information regarding the TEVs-mediated CD44s/CD44v transfer/interaction in the context of cancer metastasis. The content and the impact of the transferred cargo by this type of TEVs also are discussed with regards to tumor cell dissemination.
Collapse
|
6
|
Medrano-González PA, Rivera-Ramírez O, Montaño LF, Rendón-Huerta EP. Proteolytic Processing of CD44 and Its Implications in Cancer. Stem Cells Int 2021; 2021:6667735. [PMID: 33505471 PMCID: PMC7811561 DOI: 10.1155/2021/6667735] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/04/2020] [Accepted: 12/24/2020] [Indexed: 01/16/2023] Open
Abstract
CD44 is a transmembrane glycoprotein expressed in several healthy and tumor tissues. Modifications in its structure contribute differently to the activity of this molecule. One modification that has provoked interest is the consecutive cleavage of the CD44 extracellular ectodomain by enzymes that belong mainly to the family of metalloproteases. This process releases biologically active substrates, via alternative splice forms of CD44, that generate CD44v3 or v6 isoforms which participate in the transcriptional regulation of genes and proteins associated to signaling pathways involved in the development of cancer. These include the protooncogene tyrosine-protein kinase Src (c-Src)/signal transducer and activator of transcription 3 (STAT3), the epithelial growth factor receptor, the estrogen receptor, Wnt/βcatenin, or Hippo signaling pathways all of which are associated to cell proliferation, differentiation, or cancer progression. Whereas CD44 still remains as a very useful prognostic cell marker in different pathologies, the main topic is that the generation of CD44 intracellular fragments assists the regulation of transcriptional proteins involved in the cell cycle, cell metabolism, and most importantly, the regulation of some stem cell-associated markers.
Collapse
Affiliation(s)
- Priscila Anhel Medrano-González
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edif. D, 1 piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, 04510 Mexico, Mexico
| | - Osmar Rivera-Ramírez
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
| | - Luis Felipe Montaño
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
| | - Erika P. Rendón-Huerta
- Lab. Inmunobiología, Depto. Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico, Mexico
| |
Collapse
|
7
|
Hassan N, Greve B, Espinoza-Sánchez NA, Götte M. Cell-surface heparan sulfate proteoglycans as multifunctional integrators of signaling in cancer. Cell Signal 2020; 77:109822. [PMID: 33152440 DOI: 10.1016/j.cellsig.2020.109822] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
Proteoglycans (PGs) represent a large proportion of the components that constitute the extracellular matrix (ECM). They are a diverse group of glycoproteins characterized by a covalent link to a specific glycosaminoglycan type. As part of the ECM, heparan sulfate (HS)PGs participate in both physiological and pathological processes including cell recruitment during inflammation and the promotion of cell proliferation, adhesion and motility during development, angiogenesis, wound repair and tumor progression. A key function of HSPGs is their ability to modulate the expression and function of cytokines, chemokines, growth factors, morphogens, and adhesion molecules. This is due to their capacity to act as ligands or co-receptors for various signal-transducing receptors, affecting pathways such as FGF, VEGF, chemokines, integrins, Wnt, notch, IL-6/JAK-STAT3, and NF-κB. The activation of those pathways has been implicated in the induction, progression, and malignancy of a tumor. For many years, the study of signaling has allowed for designing specific drugs targeting these pathways for cancer treatment, with very positive results. Likewise, HSPGs have become the subject of cancer research and are increasingly recognized as important therapeutic targets. Although they have been studied in a variety of preclinical and experimental models, their mechanism of action in malignancy still needs to be more clearly defined. In this review, we discuss the role of cell-surface HSPGs as pleiotropic modulators of signaling in cancer and identify them as promising markers and targets for cancer treatment.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Biotechnology Program, Department of Chemistry, Faculty of Science, Cairo University, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany.
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.
| |
Collapse
|
8
|
Mereiter S, Martins ÁM, Gomes C, Balmaña M, Macedo JA, Polom K, Roviello F, Magalhães A, Reis CA. O‐glycan truncation enhances cancer‐related functions of
CD
44 in gastric cancer. FEBS Lett 2019; 593:1675-1689. [DOI: 10.1002/1873-3468.13432] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/04/2019] [Accepted: 05/06/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Stefan Mereiter
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Álvaro M. Martins
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Catarina Gomes
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Meritxell Balmaña
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Joana A. Macedo
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Karol Polom
- Department of Surgical Oncology Medical University of Gdansk Poland
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Franco Roviello
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Ana Magalhães
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
- Faculty of Medicine University of Porto Portugal
- Instituto de Ciências Biomédicas Abel Salazar University of Porto Portugal
| |
Collapse
|
9
|
Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix Biol 2018; 78-79:292-313. [PMID: 29625181 DOI: 10.1016/j.matbio.2018.03.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The tissue microenvironment contributes to local immunity and to the pathogenesis of autoimmune diseases - a diverse set of conditions characterized by sterile inflammation, immunity against self-antigens, and destruction of tissues. However, the specific factors within the tissue microenvironment that contribute to local immune dysregulation in autoimmunity are poorly understood. One particular tissue component implicated in multiple autoimmune diseases is hyaluronan (HA), an extracellular matrix (ECM) polymer. HA is abundant in settings of chronic inflammation and contributes to lymphocyte activation, polarization, and migration. Here, we first describe what is known about the size, amount, and distribution of HA at sites of autoimmunity and in associated lymphoid structures in type 1 diabetes, multiple sclerosis, and rheumatoid arthritis. Next, we examine the recent literature on HA and its impact on adaptive immunity, particularly in regards to the biology of lymphocytes and Foxp3+ regulatory T-cells (Treg), a T-cell subset that maintains immune tolerance in healthy individuals. We propose that HA accumulation at sites of chronic inflammation creates a permissive environment for autoimmunity, characterized by CD44-mediated inhibition of Treg expansion. Finally, we address potential tools and strategies for targeting HA and its receptor CD44 in chronic inflammation and autoimmunity.
Collapse
Affiliation(s)
- Nadine Nagy
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton L Marshall
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Esther Wang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
10
|
Dumont C, Czuba E, Chen M, Villadangos JA, Johnston AP, Mintern JD. DNA‐based probes for flow cytometry analysis of endocytosis and recycling. Traffic 2017; 18:242-249. [DOI: 10.1111/tra.12466] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Claire Dumont
- Department of Biochemistry and Molecular Biology The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute Parkville Victoria Australia
| | - Ewa Czuba
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria Australia
| | - Moore Chen
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria Australia
| | - Jose A. Villadangos
- Department of Biochemistry and Molecular Biology The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute Parkville Victoria Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity The University of Melbourne Parkville Victoria Australia
| | - Angus P.R. Johnston
- Drug Delivery, Disposition and Dynamics Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology Monash University Parkville Australia
| | - Justine D. Mintern
- Department of Biochemistry and Molecular Biology The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute Parkville Victoria Australia
| |
Collapse
|
11
|
Liu H, Dumont C, Johnston APR, Mintern JD. Analysis of Intracellular Trafficking of Dendritic Cell Receptors for Antigen Targeting. Methods Mol Biol 2016; 1423:199-209. [PMID: 27142019 DOI: 10.1007/978-1-4939-3606-9_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Antibody-targeted vaccination aims to efficiently deliver antigen to dendritic cells by targeting specific receptors at the cell surface. The choice of receptor depends on different factors, including their capacity to induce internalization of the delivered antigen/adjuvant cargo. Assays currently used to monitor internalization in dendritic cells have several limitations. We have developed a novel DNA-based probe that allows for simple and robust high-throughput analysis of internalization. Designed for flow cytometry, the probe can also be used for fluorescence microscopy to clearly distinguish internalized from surface-bound material. Here, we describe the steps for modifying material (antibodies, proteins) with the probe, undertaking the assay, and analyzing the data obtained from flow cytometry.
Collapse
Affiliation(s)
- Haiyin Liu
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC, 3010, Australia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Claire Dumont
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC, 3010, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC, 3010, Australia.
| |
Collapse
|
12
|
Hartmann M, Herrlich A, Herrlich P. Who decides when to cleave an ectodomain? Trends Biochem Sci 2013; 38:111-20. [PMID: 23298902 DOI: 10.1016/j.tibs.2012.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 11/27/2012] [Accepted: 12/03/2012] [Indexed: 10/27/2022]
Abstract
Many life-essential molecules such as growth factors, cytokines, ectoenzymes, and decoy receptors are produced by ectodomain cleavage of transmembrane precursor molecules. Not surprisingly, misregulation of such essential functions is linked to numerous diseases. Ectodomain cleavage is the function of transmembrane ADAMs (a disintegrin and metalloprotease) and other membrane-bound metalloproteases, which have an extracellular catalytic domain. Almost all work on ectodomain cleavage regulation has focused on the control of enzyme activity determined by substrate cleavage as surrogate. However, the number of substrates far exceeds the number of enzymes. Specificity can therefore not be achieved by solely modulating enzyme activity. Here, we argue that specific regulatory pathways must exist to control the availability and susceptibility of substrates.
Collapse
Affiliation(s)
- Monika Hartmann
- Leibniz Institute for Age Research - Fritz Lipmann Institute, Herrlich Laboratory, Beutenbergstr. 11, 07745 Jena, Germany
| | | | | |
Collapse
|
13
|
Du J, Liu Y, Meline B, Kong G, Tan LX, Lo JC, Wang J, Ranheim E, Zhang L, Chang YI, Ryu MJ, Zhang JF, Zhang J. Loss of CD44 attenuates aberrant GM-CSF signaling in Kras G12D hematopoietic progenitor/precursor cells and prolongs the survival of diseased animals. Leukemia 2012; 27:754-7. [PMID: 22976127 DOI: 10.1038/leu.2012.251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
14
|
Nesterovitch AB, Szanto S, Gonda A, Bardos T, Kis-Toth K, Adarichev VA, Olasz K, Ghassemi-Najad S, Hoffman MD, Tharp MD, Mikecz K, Glant TT. Spontaneous insertion of a b2 element in the ptpn6 gene drives a systemic autoinflammatory disease in mice resembling neutrophilic dermatosis in humans. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1701-14. [PMID: 21435452 DOI: 10.1016/j.ajpath.2010.12.053] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 12/15/2010] [Accepted: 12/23/2010] [Indexed: 01/01/2023]
Abstract
We found a spontaneous autosomal mutation in a mouse leading to neutrophil infiltration with ulceration in the upper dermis of homozygous offspring. These animals had increased neutrophil numbers, associated with normal lymphocyte count, in peripheral blood and bone marrow, suggesting a myeloproliferative disorder; however, granulocyte precursor proliferation in bone marrow was actually reduced (because circulating neutrophils were less susceptible to apoptosis). Neutrophil infiltration of the skin and other organs and high serum levels of immunoglobulins and autoantibodies, cytokines, and acute-phase proteins were additional abnormalities, all of which could be reduced by high-dose corticosteroid treatment or neutrophil depletion by antibodies. Use of genome-wide screening localized the mutation within an 0.4-Mbp region on mouse chromosome 6. We identified insertion of a B2 element in exon 6 of the Ptpn6 gene (protein tyrosine phosphatase, non-receptor type 6; also known as Shp-1). This insertion involves amino acid substitutions that significantly reduced the enzyme activity in mice homozygous for the mutation. Disease onset was delayed, and the clinical phenotype was milder than the phenotypes of other Ptpn6-mutants described in motheaten (me, mev) mice; we designated this new genotype as Ptpn6(meB2/meB2) and the phenotype as meB2. This new phenotype encompasses an autoinflammatory disease showing similarities to many aspects of the so-called neutrophilic dermatoses, a heterogeneous group of skin diseases with unknown etiology in humans.
Collapse
|
15
|
Deng W, Srinivasan S, Zheng X, Putkey JA, Li R. Interaction of calmodulin with L-selectin at the membrane interface: implication on the regulation of L-selectin shedding. J Mol Biol 2011; 411:220-33. [PMID: 21664913 DOI: 10.1016/j.jmb.2011.05.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/21/2011] [Accepted: 05/26/2011] [Indexed: 12/18/2022]
Abstract
The calmodulin (CaM) hypothesis of ectodomain shedding stipulates that CaM, an intracellular Ca²⁺-dependent regulatory protein, associates with the cytoplasmic domain of L-selectin to regulate ectodomain shedding of L-selectin on the other side of the plasma membrane. To understand the underlying molecular mechanism, we have characterized the interactions of CaM with two peptides derived from human L-selectin. The peptide ARR18 corresponds to the entire cytoplasmic domain of L-selectin (residues Ala317-Tyr334 in the mature protein), and CLS corresponds to residues Lys280-Tyr334, which contains the entire transmembrane and cytoplasmic domains of l-selectin. Monitoring the interaction by fluorescence spectroscopy and other biophysical techniques, we found that CaM can bind to ARR18 in aqueous solutions or the L-selectin cytoplasmic domain of CLS reconstituted in the phosphatidylcholine bilayer, both with an affinity of approximately 2 μM. The association is calcium independent and dynamic and involves both lobes of CaM. In a phospholipid bilayer, the positively charged L-selectin cytoplasmic domain of CLS is associated with anionic phosphatidylserine (PS) lipids at the membrane interface through electrostatic interactions. Under conditions where the PS content mimics that in the inner leaflet of the cell plasma membrane, the interaction between CaM and CLS becomes undetectable. These results suggest that the association of CaM with L-selectin in the cell can be influenced by the membrane bilayer and that anionic lipids may modulate ectodomain shedding of transmembrane receptors.
Collapse
Affiliation(s)
- Wei Deng
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
16
|
Nagyeri G, Radacs M, Ghassemi-Nejad S, Tryniszewska B, Olasz K, Hutas G, Gyorfy Z, Hascall VC, Glant TT, Mikecz K. TSG-6 protein, a negative regulator of inflammatory arthritis, forms a ternary complex with murine mast cell tryptases and heparin. J Biol Chem 2011; 286:23559-69. [PMID: 21566135 DOI: 10.1074/jbc.m111.222026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TSG-6 (TNF-α-stimulated gene/protein 6), a hyaluronan (HA)-binding protein, has been implicated in the negative regulation of inflammatory tissue destruction. However, little is known about the tissue/cell-specific expression of TSG-6 in inflammatory processes, due to the lack of appropriate reagents for the detection of this protein in vivo. Here, we report on the development of a highly sensitive detection system and its use in cartilage proteoglycan (aggrecan)-induced arthritis, an autoimmune murine model of rheumatoid arthritis. We found significant correlation between serum concentrations of TSG-6 and arthritis severity throughout the disease process, making TSG-6 a better biomarker of inflammation than any of the other arthritis-related cytokines measured in this study. TSG-6 was present in arthritic joint tissue extracts together with the heavy chains of inter-α-inhibitor (IαI). Whereas TSG-6 was broadly detectable in arthritic synovial tissue, the highest level of TSG-6 was co-localized with tryptases in the heparin-containing secretory granules of mast cells. In vitro, TSG-6 formed complexes with the tryptases murine mast cell protease-6 and -7 via either heparin or HA. In vivo TSG-6-tryptase association could also be detected in arthritic joint extracts by co-immunoprecipitation. TSG-6 has been reported to suppress inflammatory tissue destruction by enhancing the serine protease-inhibitory activity of IαI against plasmin. TSG-6 achieves this by transferring heavy chains from IαI to HA, thus liberating the active bikunin subunit of IαI. Because bikunin is also present in mast cell granules, we propose that TSG-6 can promote inhibition of tryptase activity via a mechanism similar to inhibition of plasmin.
Collapse
Affiliation(s)
- Gyorgy Nagyeri
- Section of Molecular Medicine, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Hayashida K, Bartlett AH, Chen Y, Park PW. Molecular and cellular mechanisms of ectodomain shedding. Anat Rec (Hoboken) 2010; 293:925-37. [PMID: 20503387 DOI: 10.1002/ar.20757] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The extracellular domain of several membrane-anchored proteins is released from the cell surface as soluble proteins through a regulated proteolytic mechanism called ectodomain shedding. Cells use ectodomain shedding to actively regulate the expression and function of surface molecules, and modulate a wide variety of cellular and physiological processes. Ectodomain shedding rapidly converts membrane-associated proteins into soluble effectors and, at the same time, rapidly reduces the level of cell surface expression. For some proteins, ectodomain shedding is also a prerequisite for intramembrane proteolysis, which liberates the cytoplasmic domain of the affected molecule and associated signaling factors to regulate transcription. Ectodomain shedding is a process that is highly regulated by specific agonists, antagonists, and intracellular signaling pathways. Moreover, only about 2% of cell surface proteins are released from the surface by ectodomain shedding, indicating that cells selectively shed their protein ectodomains. This review will describe the molecular and cellular mechanisms of ectodomain shedding, and discuss its major functions in lung development and disease.
Collapse
Affiliation(s)
- Kazutaka Hayashida
- Division of Respiratory Diseases, Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
18
|
Wiranowska M, Ladd S, Moscinski LC, Hill B, Haller E, Mikecz K, Plaas A. Modulation of hyaluronan production by CD44 positive glioma cells. Int J Cancer 2010; 127:532-42. [PMID: 19957333 PMCID: PMC3962665 DOI: 10.1002/ijc.25085] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study examines the functional relationship between glioma cell production of hyaluronan (HA), known to play a role in glioma invasion, expression of its CD44 receptor, and glioma cell viability. Production of HA by CD44 positive mouse G26 and human U373 glioma cell lines was evaluated and compared to that of a CD44 positive mouse fibroblast-like L929 cell line. We found that both G26 and U373 MG glioma cells, but not L929 fibroblast-like cells, synthesized HA. The synthesis of HA by glioma cells was found during the proliferative phase as well as post-confluency, as detected by fluorophore-assisted carbohydrate electrophoresis. Eighty to ninety percent of the HA synthesized was secreted into the medium and 10-20% remained associated with the cells. To examine a possible mechanistic link between the CD44-HA interaction and endogenous HA production, glioma cells were treated with either anti-CD44 antibodies (clones KM201 or IM7) or HA oligosaccharides (hexamer oligoHA-6 or decamer oligoHA-10). We found that oligoHA-10, which was previously shown to compete effectively with the CD44-HA interaction, enhanced glioma HA synthesis by approximately 1.5-fold, without affecting cell viability. IM7 treatment of human U373 glioma cells resulted in over 50% decrease of HA production, which was associated with changes in cell size and apoptosis. Taken together, these data show that CD44 specific ligands, such as the IM7 antibody or oligoHA-10 could down-regulate or up-regulate glioma HA production, respectively. Our results suggest that interference with CD44/HA may lead to the discovery and development of new treatment modalities for glioma.
Collapse
Affiliation(s)
- Marzenna Wiranowska
- Department of Pathology and Cell Biology, University of South Florida, College of Medicine, Tampa, FL 33612-4799, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Differentially regulated GPVI ectodomain shedding by multiple platelet-expressed proteinases. Blood 2010; 116:3347-55. [PMID: 20644114 DOI: 10.1182/blood-2010-06-289108] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Glycoprotein VI (GPVI) mediates platelet activation on exposed subendothelial collagens at sites of vascular injury and thereby contributes to normal hemostasis, but also to the occlusion of diseased vessels in the setting of myocardial infarction or stroke. GPVI is an attractive target for antithrombotic therapy, particularly because previous studies have shown that anti-GPVI antibodies induce irreversible down-regulation of the receptor in circulating platelets by internalization and/or ectodomain shedding. Metalloproteinases of the a disintegrin and metalloproteinase (ADAM) family have been proposed to mediate this ectodomain shedding, but direct evidence for this is lacking. Here, we studied GPVI shedding in vitro and in vivo in newly generated mice with a megakaryocyte-specific ADAM10 deficiency and in Adam17(ex/ex) mice, which lack functional ADAM17. We demonstrate that GPVI cleavage in vitro can occur independently through either ADAM10 or ADAM17 in response to distinct stimuli. In contrast, antibody (JAQ1)-induced GPVI shedding in vivo occurred in mice lacking both ADAM10/ADAM17 in their platelets, suggesting the existence of a third GPVI cleaving platelet enzyme. This was supported by in vitro studies on ADAM10/ADAM17 double-deficient platelets. These results reveal that ectodomain shedding of GPVI can be mediated through multiple differentially regulated platelet-expressed proteinases with obvious therapeutic implications.
Collapse
|
20
|
Runnels HA, Weber GL, Min J, Kudlacz EM, Zobel JF, Donovan CB, Thiede MA, Zhang J, Alpert RB, Salafia MA, Milici AJ, Burdette D, Bell RR, Beebe JS, Xu X. PF-03475952: a potent and neutralizing fully human anti-CD44 antibody for therapeutic applications in inflammatory diseases. Adv Ther 2010; 27:168-80. [PMID: 20429046 DOI: 10.1007/s12325-010-0010-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Indexed: 12/11/2022]
Abstract
INTRODUCTION CD44 is a cell adhesion molecule believed to play a critical role in T cell and monocyte infiltration in the inflammatory process. The reduction of CD44 expression or its ability to properly interact with its key ligand, hyaluronic acid (HA), inhibits migration and subsequent activation of cells within sites of inflammation. CD44-deficient mice exhibit decreased disease in a mouse arthritis model. METHODS Accordingly, we developed PF-03475952, a fully human IgG2 anti-CD44 monoclonal antibody (mAb). RESULTS Binding of PF-03475952 to CD44 inhibits binding of HA and induces loss of CD44 from the cell surface. PF-03475952 also passed a series of safety pharmacology assays designed to assess the risk of the mAb to bind Fc gamma receptors, stimulate cytokine release from human whole blood, and stimulate cytokine release from peripheral blood mononuclear cells (PBMC) using plate-bound antibodies. The latter assay was designed specifically to evaluate the risk of cytokine storm that had been observed with TGN1412 (immunostimulatory CD28 superagonist mAb). PF-003475952 exhibits high-affinity binding to both human and cynomolgus monkey CD44, but does not cross-react with rodent CD44. Thus, a rat anti-mouse CD44 mAb was used to demonstrate a dose-dependent decrease of disease in mouse collagen-induced arthritis. Importantly, efficacy was correlated with >50% loss of cell surface CD44 on circulating cells. Loss of CD44 expression on CD3+ lymphocytes was monitored following a single dose of PF-03475952 in cynomolgus monkeys as a pharmacodynamic marker. The recovery of CD44 expression was found to be dose-dependent. PF-03475952 doses of 1, 10, and 100 mg/kg reduced CD44 expression below 50% for 218, 373, and >504 hours, respectively. CONCLUSION Targeting of CD44 is a unique mechanism of action in the treatment of inflammatory diseases and is expected to reduce joint damage induced by inflammatory mediators, resulting in disease modification in inflammatory diseases such as rheumatoid arthritis.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neutralizing/pharmacology
- Antibodies, Neutralizing/therapeutic use
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Cytokines/blood
- Enzyme-Linked Immunosorbent Assay
- Humans
- Hyaluronan Receptors/immunology
- Hyaluronan Receptors/metabolism
- Hyaluronic Acid/metabolism
- Immunoglobulin G/pharmacology
- Immunoglobulin G/therapeutic use
- Macaca fascicularis
- Male
- Mice
- Mice, Inbred DBA
- Platelet Activation/drug effects
- Protein Binding
Collapse
Affiliation(s)
- Herbert A Runnels
- Pfizer Global Research and Development, St. Louis Laboratories, Chesterfield, MO 63017, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Duterme C, Mertens-Strijthagen J, Tammi M, Flamion B. Two novel functions of hyaluronidase-2 (Hyal2) are formation of the glycocalyx and control of CD44-ERM interactions. J Biol Chem 2009; 284:33495-508. [PMID: 19783662 DOI: 10.1074/jbc.m109.044362] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It has long been predicted that the members of the hyaluronidase enzyme family have important non-enzymatic functions. However, their nature remains a mystery. The metabolism of hyaluronan (HA), their major enzymatic substrate, is also enigmatic. To examine the function of Hyal2, a glycosylphosphatidylinositol-anchored hyaluronidase with intrinsically weak enzymatic activity, we have compared stably transfected rat fibroblastic BB16 cell lines with various levels of expression of Hyal2. These cell lines continue to express exclusively the standard form (CD44s) of the main HA receptor, CD44. Hyal2, CD44, and one of its main intracellular partners, ezrin-radixin-moesin (ERM), were found to co-immunoprecipitate. Functionally, Hyal2 overexpression was linked to loss of the glycocalyx, the HA-rich pericellular coat. This effect could be mimicked by exposure of BB16 cells either to Streptomyces hyaluronidase, to HA synthesis inhibitors, or to HA oligosaccharides. This led to shedding of CD44, separation of CD44 from ERM, reduction in baseline level of ERM activation, and markedly decreased cell motility (50% reduction in a wound healing assay). The effects of Hyal2 on the pericellular coat and on CD44-ERM interactions were inhibited by treatment with the Na(+)/H(+) exchanger-1 inhibitor ethyl-N-isopropylamiloride. We surmise that Hyal2, through direct interactions with CD44 and possibly some pericellular hyaluronidase activity requiring acidic foci, suppresses the formation or the stability of the glycocalyx, modulates ERM-related cytoskeletal interactions, and diminishes cell motility. These effects may be relevant to the purported in vivo tumor-suppressive activity of Hyal2.
Collapse
Affiliation(s)
- Cecile Duterme
- Molecular Physiology Research Unit, University of Namur, 5000 Namur, Belgium
| | | | | | | |
Collapse
|
22
|
Chen C, Chang MC, Hsieh RK, Chang YF, Lin J, Tsan KW. Activation of CD44 facilitates DNA repair in T-cell lymphoma but has differential effects on apoptosis induced by chemotherapeutic agents and ionizing radiation. Leuk Lymphoma 2009; 46:1785-95. [PMID: 16263582 DOI: 10.1080/10428190500232501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Expression of CD44s (standard form) in malignant lymphoma is a poor indicator of survival. To investigate whether activation of CD44s can protect from cell death, this study compared the extent of apoptosis induced by chemotherapeutic agents and ionizing radiation (IR) on T-lymphoma cell lines in the presence or absence of adherent hyaluronan and monoclonal antibodies (MoAbs). Growth in the presence of adherent ligands enhanced apoptosis induced by dexamethasone (Dex), but protected cells from epirubicin-induced apoptosis. In IR-induced apoptosis, mouse lymphoma cells had resistance against apoptosis when treated with hyaluronan (HA), although acute cell death reached the same plateau regardless of treatment with adherent MoAbs in human lymphoma cell line. However, the post-irradiated repopulation of lymphoma cells was strikingly accelerated in those treated with CD44 adherent ligands. This repopulation process correlated with the remarkable upregulation of proliferating cell nuclear antigen (PCNA), which is a protein involved in DNA repair. Unscheduled DNA synthesis (UDS), a measure of DNA repair, was consistently enhanced in CD44s-stimulated cells after exposure to radiation. The results suggest that the poor prognostic indication of CD44 expression is more a consequence of enhanced DNA repair following genotoxic damage than of direct resistance to apoptosis.
Collapse
Affiliation(s)
- C Chen
- Mackay Medicine, Nursing, and Management College, Taipei, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
23
|
Pruessmeyer J, Ludwig A. The good, the bad and the ugly substrates for ADAM10 and ADAM17 in brain pathology, inflammation and cancer. Semin Cell Dev Biol 2009; 20:164-74. [DOI: 10.1016/j.semcdb.2008.09.005] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 09/11/2008] [Accepted: 09/15/2008] [Indexed: 10/21/2022]
|
24
|
Anderegg U, Eichenberg T, Parthaune T, Haiduk C, Saalbach A, Milkova L, Ludwig A, Grosche J, Averbeck M, Gebhardt C, Voelcker V, Sleeman JP, Simon JC. ADAM10 is the constitutive functional sheddase of CD44 in human melanoma cells. J Invest Dermatol 2008; 129:1471-82. [PMID: 18971959 DOI: 10.1038/jid.2008.323] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
CD44 proteins are cell surface receptors for hyaluronic acid (HA), a component of the extracellular matrix that has multiple effects on cell behavior. CD44 can be shed from the cell surface by proteolytic cleavage. The resulting soluble form can interfere with the interaction between HA and membrane-bound CD44. Soluble CD44 can abolish the cell proliferation-promoting effect of HA on melanoma cell lines, suggesting that a better understanding of the shedding process might identify ways of blocking tumor cell proliferation. ADAM10, ADAM17, and MMP14 have previously been implicated in the shedding of CD44 from various tumor cells. Using immunohistochemistry we demonstrate that ADAM10 and ADAM17 but not MMP14 are significantly expressed on melanoma cells in histological sections. In human melanoma cell lines expression of these proteases could be blocked by transfection with appropriate siRNAs. However, only blocking of ADAM10 expression led to decreased shedding of CD44. In parallel, cell proliferation was promoted. Confocal microscopy demonstrated that ADAM10 and CD44 colocalize on the cell surface. We conclude that ADAM10 is the predominant protease involved in the constitutive shedding of endogenous CD44 from melanoma cells, and that enhancement of ADAM10 activity could be an approach to decrease the proliferation of melanoma cells.
Collapse
Affiliation(s)
- Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Borg N, Holland M. The effect of glycosaminoglycans on rat gametes in vitro and the associated signal pathway. Reproduction 2008; 135:311-9. [DOI: 10.1530/rep-07-0267] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The effects of adding the extracellular glycosaminoglycans (GAGs), hyaluronic acid (HA) and chondroitin sulphate (CS) to ratin vitrofertilisation (IVF) media were assessed. Metaphase II (MII) oocytes were also incubated in GAG-supplemented modified rat 1-cell embryo culture medium (mR1ECM+BSA) for 3 days. Cytoplasmic fragmentation was significantly reduced in mR1ECM+BSA with HA (39.0–48.0%) compared with the control (82.0%). In IVF experiments, neither HA (8.0–30.8%) nor CS (9.7–42.5%) improved fertilisation rates compared with controls fertilised in M16 (47.2%) or enriched Krebs–Ringer bicarbonate solution (61.5%). RT-PCR and Western blot were used to probe for CD44 mRNA and protein in Sprague–Dawley gametes and cumulus cells. CD44 was identified in cumulus cells, suggesting a role for oocyte maturation and cumulus expansion. The CD44 protein was also present on caudal epididymal spermatozoa that were highly stimulated by CSin vitroimplicating a role in fertilisation for CS and CD44.
Collapse
|
26
|
Roda-Navarro P, Reyburn HT. Intercellular protein transfer at the NK cell immune synapse: mechanisms and physiological significance. FASEB J 2007; 21:1636-46. [PMID: 17314139 DOI: 10.1096/fj.06-7488rev] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immune synapses (IS) are supramolecular clusters providing intercellular communication among cells of the immune system. While the physiological role and consequences of IS formation are beginning to be understood, these studies have given rise to a new research topic in the biology of lymphocyte interactions: synaptic transfer of proteins between lymphocytes. During natural killer (NK) cell immunosurveillance, clustering and transfer of receptor and ligand molecules have been observed at both the inhibitory and cytotoxic NK cell immune synapse (NK-IS). The transfer of activating receptors seems to be associated with receptor distribution to thin membrane connective structures (MCS)/nanotubes that communicate effector and susceptible target cells. Strikingly, bidirectional transfer of the activating receptor NKG2D and its cellular ligand MICB correlates with a reduction in NK cell cytotoxic function. In this regard, synaptic uptake of MICB may represent a novel strategy of tumor immune evasion. Finally, synaptic acquisition of receptors by NK cells may also favor the spread of pathogens. In this review we discuss possible mechanisms of synaptic protein transfer and propose different testable hypotheses about the physiological and pathological significance of this process for NK cell function.
Collapse
|
27
|
Wang JHC, Thampatty BP, Lin JS, Im HJ. Mechanoregulation of gene expression in fibroblasts. Gene 2007; 391:1-15. [PMID: 17331678 PMCID: PMC2893340 DOI: 10.1016/j.gene.2007.01.014] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 01/08/2007] [Accepted: 01/15/2007] [Indexed: 12/11/2022]
Abstract
Mechanical loads placed on connective tissues alter gene expression in fibroblasts through mechanotransduction mechanisms by which cells convert mechanical signals into cellular biological events, such as gene expression of extracellular matrix components (e.g., collagen). This mechanical regulation of ECM gene expression affords maintenance of connective tissue homeostasis. However, mechanical loads can also interfere with homeostatic cellular gene expression and consequently cause the pathogenesis of connective tissue diseases such as tendinopathy and osteoarthritis. Therefore, the regulation of gene expression by mechanical loads is closely related to connective tissue physiology and pathology. This article reviews the effects of various mechanical loading conditions on gene regulation in fibroblasts and discusses several mechanotransduction mechanisms. Future research directions in mechanoregulation of gene expression are also suggested.
Collapse
Affiliation(s)
- James H-C Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, 210 Lothrop St., BST, E1640, Pittsburgh, PA 15213, USA.
| | | | | | | |
Collapse
|
28
|
Sugahara KN, Hirata T, Hayasaka H, Stern R, Murai T, Miyasaka M. Tumor cells enhance their own CD44 cleavage and motility by generating hyaluronan fragments. J Biol Chem 2005; 281:5861-8. [PMID: 16407205 DOI: 10.1074/jbc.m506740200] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyaluronan (HA) is an extracellular matrix glycosaminoglycan that interacts with cell-surface receptors, including CD44. Although HA usually exists as a high molecular mass polymer, HA of a much lower molecular mass that shows a variety of biological activities can be detected under certain pathological conditions, particularly in tumors. We previously reported that low molecular weight HAs (LMW-HAs) of a certain size range induce the proteolytic cleavage of CD44 from the surface of tumor cells and promote tumor cell migration in a CD44-dependent manner. Here, we show that MIA PaCa-2, a human pancreatic carcinoma cell line, secreted hyaluronidases abundantly and generated readily detectable levels of LMW-HAs ranging from approximately 10- to 40-mers. This occurred in the absence of any exogenous stimulation. The tumor-derived HA oligosaccharides were able to enhance CD44 cleavage and tumor cell motility. Inhibition of the CD44-HA interaction resulted in the complete abrogation of these cellular events. These results are consistent with the concept that tumor cells generate HA oligosaccha-rides that bind to tumor cell CD44 through the expression of their own constitutive hyaluronidases. This enhances their own CD44 cleavage and cell motility, which would subsequently promote tumor progression. Such an autocrine/paracrine-like process may represent a novel activation mechanism that would facilitate and promote the malignant potential of tumor cells.
Collapse
Affiliation(s)
- Kazuki N Sugahara
- Laboratory of Immunodynamics, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Ahram M, Strittmatter EF, Monroe ME, Adkins JN, Hunter JC, Miller JH, Springer DL. Identification of shed proteins from Chinese hamster ovary cells: application of statistical confidence using human and mouse protein databases. Proteomics 2005; 5:1815-26. [PMID: 15815987 DOI: 10.1002/pmic.200401072] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The shedding process releases ligands, receptors, and other proteins from the surface of the cell and is a mechanism whereby cells communicate. Even though altered regulation of this process has been implicated in several diseases, global approaches to evaluate shed proteins have not been developed. A goal of this study was to identify global changes in shed proteins in media taken from cells exposed to low-doses of radiation to develop a fundamental understanding of the bystander response. Chinese hamster ovary cells were chosen because they have been widely used for radiation studies and are reported to respond to radiation by releasing factors into the media that cause genomic instability and cytotoxicity in unexposed cells, i.e., a bystander effect. Media samples taken for irradiated cells were evaluated using a combination of tandem- and Fourier transform-ion cyclotron resonance (FT-ICR)-mass spectrometry (MS) analyses. Since the hamster genome has not been sequenced, MS data was searched against the mouse and human protein databases. Nearly 150 proteins identified by tandem mass spectrometry were confirmed by FT-ICR. When both types of MS data were evaluated, using a new confidence scoring tool based on discriminant analyses, about 500 proteins were identified. Approximately 20% of these identifications were either integral membrane proteins or membrane associated proteins, suggesting that they were derived from the cell surface and, hence were likely shed. However, estimates of quantitative changes, based on two independent MS approaches, did not identify any protein abundance changes attributable to the bystander effect. Results from this study demonstrate the feasibility of global evaluation of shed proteins using MS in conjunction with cross-species protein databases and that significant improvement in peptide/protein identifications is provided by the confidence scoring tool.
Collapse
Affiliation(s)
- Mamoun Ahram
- Biological Sciences Division, Battelle, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Annabi B, Bouzeghrane M, Currie JC, Hawkins R, Dulude H, Daigneault L, Ruiz M, Wisniewski J, Garde S, Rabbani SA, Panchal C, Wu JJ, Béliveau R. A PSP94-derived Peptide PCK3145 inhibits MMP-9 Secretion and Triggers CD44 Cell Surface Shedding: Implication in Tumor Metastasis. Clin Exp Metastasis 2005; 22:429-39. [PMID: 16283486 DOI: 10.1007/s10585-005-2669-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Accepted: 09/01/2005] [Indexed: 10/25/2022]
Abstract
PURPOSE PCK3145 is a synthetic peptide corresponding to amino acids 31-45 of prostate secretory protein 94, which can reduce experimental skeletal metastases and prostate tumor growth in vivo. Part of its biological action involves the reduction of circulating plasma matrix metalloproteinase (MMP)-9, a crucial mediator in extracellular matrix (ECM) degradation during tumor metastasis and cancer cell invasion. The antimetastatic mechanism of action of PCK3145 is however, not understood. EXPERIMENTAL DESIGN HT-1080 fibrosarcoma cells were treated with PCK3145, and cell lysates used for immunoblot analysis of small GTPase RhoA and membrane type (MT)1-MMP protein expression. Conditioned media was used to monitor soluble MMP-9 gelatinolytic activity by zymography and protein expression by immunoblotting. RT-PCR was used to assess RhoA, MT1-MMP, MMP-9, RECK, and CD44 gene expression. Flow cytometry was used to monitor cell surface expression of CD44 and of membrane-bound MMP-9. Cell adhesion was performed on different purified ECM proteins, while cell migration was specifically performed on hyaluronic acid (HA). RESULTS We found that PCK3145 inhibited HT-1080 cell adhesion onto HA, laminin-1, and type-I collagen suggesting the common implication of the cell surface receptor CD44. In fact, PCK3145 triggered the shedding of CD44 from the cell surface into the conditioned media. PCK3145 also inhibited MMP-9 secretion and binding to the cell surface. This effect was correlated to increased RhoA and MT1-MMP gene and protein expression. CONCLUSIONS Our data suggest that PCK3145 may antagonize tumor cell metastatic processes by inhibiting both MMP-9 secretion and its potential binding to its cell surface docking receptor CD44. Such mechanism may involve RhoA signaling and increase in MT1-MMP-mediated CD44 shedding. Together with its beneficial effects in clinical trials, this is the first demonstration of PCK3145 acting as a MMP secretion inhibitor.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie-Biochimie, Université du Québec à Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ahram M, Adkins JN, Auberry DL, Wunschel DS, Springer DL. A proteomic approach to characterize protein shedding. Proteomics 2005; 5:123-31. [PMID: 15672459 DOI: 10.1002/pmic.200400912] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Shedding (i.e. proteolysis of ectodomains of membrane proteins) plays an important pathophysiological role. In order to study the feasibility of identifying shed proteins, we analyzed serum-free media of human mammary epithelial cells by mass spectrometry following induction of shedding by the phorbol ester, 4 beta-phorbol 12-myristate 13-acetate (PMA). Different means of sample preparation, including biotinylation of cell surface proteins, isolation of glycosylated proteins, and preparation of crude protein fractions, were carried out to develop the optimal method of sample processing. The collected proteins were digested with trypsin and analyzed by reversed-phase capillary liquid chromatography interfaced to an ion-trap mass spectrometer. The resulting peptide spectra were interpreted using the program SEQUEST. Analyzing the sample containing the crude protein mixture without chemical modification or separation resulted in the greatest number of identifications, including putatively shed proteins. Overall, 45 membrane-associated proteins were identified including 22 that contain at least one transmembrane domain and 23 that indirectly associate with the extracellular surface of the plasma membrane. Of the 22 transmembrane proteins, 18 were identified by extracellular peptides providing strong evidence they originate from regulated proteolysis or shedding processes. We combined results from the different experiments and used a peptide count method to estimate changes in protein abundance. Using this approach, we identified two proteins, syndecan-4 and hepatoma-derived growth factor, whose abundances increased in media of cells treated with PMA. We also detected proteins whose abundances decreased after PMA treatment such as 78 kDa glucose-regulated protein and lactate dehydrogenase A. Further analysis using immunoblotting validated the abundance changes for syndecan-4 and 78 kDa glucose-regulated protein as a result of PMA treatment. These results demonstrate that tandem mass spectrometry can be used to identify shed proteins and to estimate changes in protein abundance.
Collapse
Affiliation(s)
- Mamoun Ahram
- Biological Sciences Division, Battelle, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | | | | | | | | |
Collapse
|
32
|
Bohlson SS, Silva R, Fonseca MI, Tenner AJ. CD93 is rapidly shed from the surface of human myeloid cells and the soluble form is detected in human plasma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 175:1239-47. [PMID: 16002728 DOI: 10.4049/jimmunol.175.2.1239] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
CD93 is a highly glycosylated transmembrane protein expressed on monocytes, neutrophils, endothelial cells, and stem cells. Antibodies directed at CD93 modulate phagocytosis, and CD93-deficient mice are defective in the clearance of apoptotic cells from the inflamed peritoneum. In this study we observe that CD93, expressed on human monocytes and neutrophils, is susceptible to phorbol dibutyrate-induced protein ectodomain shedding in a time- and dose-dependent manner. The soluble fragment found in culture supernatant retains the N-terminal carbohydrate recognition domain and the epidermal growth factor repeats after ectodomain cleavage. Importantly, a soluble form of the CD93 ectodomain was detected in human plasma, demonstrating that shedding is a physiologically relevant process. Inhibition of metalloproteinases with 1,10-phenanthroline inhibited shedding, but shedding was independent of TNF-alpha-converting enzyme (a disintegrin and metalloproteinase 17). Phorbol dibutyrate-induced CD93 shedding on monocytes was accompanied by decreased surface expression, whereas neutrophils displayed an increase in surface expression, suggesting that CD93 shed from the neutrophil surface was rapidly replaced by CD93 from intracellular stores. Cross-linking CD93 on human monocytes with immobilized anti-CD93 mAbs triggered shedding, as demonstrated by a decrease in cell-associated, full-length CD93 concomitant with an increase in CD93 intracellular domain-containing cleavage products. In addition, the inflammatory mediators, TNF-alpha and LPS, stimulated ectodomain cleavage of CD93 from monocytes. These data demonstrate that CD93 is susceptible to ectodomain shedding, identify multiple stimuli that trigger shedding, and identify both a soluble form of CD93 in human plasma and intracellular domain containing cleavage products within cells that may contribute to the physiologic role of CD93.
Collapse
Affiliation(s)
- Suzanne S Bohlson
- Department of Molecular Biology and Biochemistry, Center for Immunology, University of California-Irvine, 2419 McGaugh Hall, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
33
|
Annabi B, Bouzeghrane M, Moumdjian R, Moghrabi A, Béliveau R. Probing the infiltrating character of brain tumors: inhibition of RhoA/ROK-mediated CD44 cell surface shedding from glioma cells by the green tea catechin EGCg. J Neurochem 2005; 94:906-16. [PMID: 15992376 DOI: 10.1111/j.1471-4159.2005.03256.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glioma cell-surface binding to hyaluronan (HA), a major constituent of the brain extracellular matrix (ECM) environment, is regulated through a complex membrane type-1 matrix metalloproteinase (MT1-MMP)/CD44/caveolin interaction that takes place at the leading edges of invading cells. In the present study, intracellular transduction pathways required for the HA-mediated recognition by infiltrating glioma cells in brain was investigated. We show that the overexpression of the GTPase RhoA up-regulated MT1-MMP expression and triggered CD44 shedding from the U-87 glioma cell surface. This potential implication in cerebral metastatic processes was also observed in cells overexpressing the full-length recombinant MT1-MMP, while the overexpression of a cytoplasmic domain truncated from of MT1-MMP failed to do so. This suggests that the cytoplasmic domain of MT1-MMP transduces intracellular signaling leading to RhoA-mediated CD44 shedding. Treatment of glioma cells with the Rho-kinase (ROK) inhibitor Y27632, or with EGCg, a green tea catechin with anti-MMP and anti-angiogenesis activities, antagonized both RhoA- and MT1-MMP-induced CD44 shedding. Conversely, overexpression of recombinant ROK stimulated CD44 release. Taken together, our results suggest that RhoA/ROK intracellular signaling regulates MT1-MMP-mediated CD44 recognition of HA. These molecular processes may partly explain the diffuse brain-infiltrating character of glioma cells within the surrounding parenchyma and thus be a target for new approaches to anti-tumor therapy.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Département de Chimie-Biochimie, Université du Québec à Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
34
|
Kirfel G, Rigort A, Borm B, Herzog V. Cell migration: mechanisms of rear detachment and the formation of migration tracks. Eur J Cell Biol 2005; 83:717-24. [PMID: 15679116 DOI: 10.1078/0171-9335-00421] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cell migration is central to many biological and pathological processes, including embryogenesis, tissue repair and regeneration as well as cancer and the inflammatory response. In general, cell migration can be usefully conceptualized as a cyclic process. The initial response of a cell to a migration-promoting agent is to polarize and extend protrusions in the direction of migration. These protrusions can be large, broad lamellipodia or spike-like filopodia, are usually driven by actin polymerization, and are stabilized by adhering to the extracellular matrix (ECM) via transmembrane receptors of the integrin family linked to the actin cytoskeleton. These adhesions serve as traction sites for migration as the cell moves forward over them, and they must be disassembled at the cell rear, allowing it to detach. The mechanisms of rear detachment and the regulatory processes involved are not well understood. The disassembly of adhesions that is required for detachment depends on a coordinated interaction of actin and actin-binding proteins, signaling molecules and effector enzymes including proteases, kinases and phosphatases. Originally, the biochemically regulated processes leading to rear detachment of migrating cells were thought not to be necessarily accompanied by any loss of cell material. However, it has been shown that during rear detachment long tubular extensions, the retracting fibers, are formed and that "membrane ripping" occurs at the cell rear. By this process, a major fraction of integrin-containing cellular material is left behind forming characteristic migration tracks that exactly mark the way a cell has taken.
Collapse
Affiliation(s)
- Gregor Kirfel
- Institute of Cell Biology, University of Bonn, Bonn, Germany.
| | | | | | | |
Collapse
|
35
|
Serobyan N, Schraufstatter IU, Strongin A, Khaldoyanidi SK. Nicotinic acetylcholine receptor-mediated stimulation of endothelial cells results in the arrest of haematopoietic progenitor cells on endothelium. Br J Haematol 2005; 129:257-65. [PMID: 15813854 DOI: 10.1111/j.1365-2141.2005.05446.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The function of endothelial cells that contribute to the regulation of haematopoietic stem/progenitor cells (HSPC) migration from peripheral blood into bone marrow can be influenced by extrinsic factors including nicotine. Therefore, the effect of nicotine on HSPC extravasation was studied. Using a parallel laminar flow chamber, we demonstrated an increase in the number of HSPC adhering to the nicotine-exposed endothelium under conditions of physiological shear stress in vitro. Nicotine-induced adhesion of HSPC was inhibited by mecamylamine, a non-selective nicotinic acetylcholine receptor (nAchR) antagonist. The enhanced adhesive interactions of HSPC with nicotine-exposed endothelial monolayers coincided with the nicotine-induced activation of endothelial cells. Nicotine induced fast cytoskeletal reorganization and formation of filopodia in endothelial cells through interaction with the non-neuronal nAchR expressed by these cells. In addition, nicotine treatment stimulated rapid phosphorylation of Erk1/2 and p-38 in endothelial cells. Finally, nicotine inhibited the stroma derived factor-1-mediated transendothelial migration of HSPC. Decreased migration of HSPC correlated with diminished matrix metalloproteinase-9 activity secreted by bone marrow cells and decreased expression of CD44 on the surface of endothelial cells. Overall, our data suggest that exposure to nicotine causes endothelial cell dysfunction and leads to the pathological arrest of HSPC on endothelium, interfering with their proper migration process.
Collapse
Affiliation(s)
- Naira Serobyan
- Division of Vascular Biology, La Jolla Institute for Molecular Medicine, San Diego, CA 92121, USA
| | | | | | | |
Collapse
|
36
|
McKallip RJ, Fisher M, Gunthert U, Szakal AK, Nagarkatti PS, Nagarkatti M. Role of CD44 and its v7 isoform in staphylococcal enterotoxin B-induced toxic shock: CD44 deficiency on hepatic mononuclear cells leads to reduced activation-induced apoptosis that results in increased liver damage. Infect Immun 2005; 73:50-61. [PMID: 15618140 PMCID: PMC538933 DOI: 10.1128/iai.73.1.50-61.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Exposure to bacterial superantigens such as staphylococcal enterotoxin B (SEB) leads to the induction of toxic shock syndrome which results in multiorgan failure, including liver damage. In the present study, we investigated the role of CD44 in SEB-induced liver injury. Injection of SEB into d-galactosamine-sensitized CD44 wild-type (WT) mice led to a significant increase in CD44 expression on liver T cells, NK cells, and NKT cells. Administration of SEB to CD44 knockout (KO) mice caused significantly enhanced liver damage which correlated with elevated numbers of T cells, NK cells, NKT cells, and macrophages in the liver and increased production of tumor necrosis factor alpha and gamma interferon compared to CD44 WT mice. Furthermore, liver mononuclear cells from CD44 KO mice were resistant to SEB-induced apoptosis, and cDNA microarray analysis revealed that SEB activation of such cells led to the induction of several antiapoptotic genes and repression of proapoptotic genes. Examination of CD44 isoforms revealed that SEB exposure altered CD44 variant 7 (v7) isoform expression. Interestingly, mice bearing a specific deletion of the CD44v7 exon exhibited increased susceptibility to SEB-induced hepatitis. Finally, treatment of CD44 WT mice with anti-CD44 monoclonal antibodies reduced expression of CD44 in liver mononuclear cells and caused increased susceptibility to SEB-induced liver injury. Together, these data demonstrate that the expression of CD44 and/or CD44v7 on SEB-activated liver mononuclear cells facilitates their rapid apoptosis, thereby preventing severe liver injury in wild-type mice, and suggest that CD44 plays an important role in the regulation and elimination of immune cells in the liver.
Collapse
Affiliation(s)
- Robert J McKallip
- Department of Microbiology and Immunology, Massey Cancer Center, Virginia Commonwealth University Medical Center, Box 980678, Richmond, VA 23298-0678.
| | | | | | | | | | | |
Collapse
|
37
|
Szántó S, Gál I, Gonda A, Glant TT, Mikecz K. Expression of L-selectin, but not CD44, is required for early neutrophil extravasation in antigen-induced arthritis. THE JOURNAL OF IMMUNOLOGY 2004; 172:6723-34. [PMID: 15153489 DOI: 10.4049/jimmunol.172.11.6723] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
L (leukocyte)-selectin (CD62L) and CD44 are major adhesion receptors that support the rolling of leukocytes on endothelium, the first step of leukocyte entry into inflamed tissue. The specific contribution of L-selectin or CD44 to the regulation of cell traffic to joints in arthritis has not been investigated. We used CD44-deficient, L-selectin-deficient, and CD44/L-selectin double knockout mice to determine the requirement for these receptors for inflammatory cell recruitment during Ag-induced arthritis. Intraperitoneal immunization resulted in similar activation status and Ag-specific responses in wild-type and gene-targeted mice. However, extravasation of neutrophil granulocytes, but not the emigration of T cells, into the knee joints after intra-articular Ag injection was significantly delayed in L-selectin-deficient and double knockout mice. Intravital videomicroscopy on the synovial microcirculation revealed enhanced leukocyte rolling and diminished adherence in mice lacking either CD44 or L-selectin, but CD44 deficiency had no significant effect on the recruitment of L-selectin-null cells. Compared with wild-type leukocytes, expression of L-selectin was down-regulated in CD44-deficient cells in the spleen, peripheral blood, and inflamed joints, suggesting that reduced expression of L-selectin, rather than the lack of CD44, could be responsible for the delayed influx of granulocytes into the joints of CD44-deficient mice. In conclusion, there is a greater requirement for L-selectin than for CD44 for neutrophil extravasation during the early phase of Ag-induced arthritis.
Collapse
Affiliation(s)
- Sándor Szántó
- Section of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
38
|
Murai T, Miyazaki Y, Nishinakamura H, Sugahara KN, Miyauchi T, Sako Y, Yanagida T, Miyasaka M. Engagement of CD44 Promotes Rac Activation and CD44 Cleavage during Tumor Cell Migration. J Biol Chem 2004; 279:4541-50. [PMID: 14623895 DOI: 10.1074/jbc.m307356200] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CD44 is a major cell surface adhesion molecule for hyaluronan, a component of the extracellular matrix, and is implicated in tumor metastasis and invasion. We reported previously that hyaluronan oligosaccharides induce CD44 cleavage from tumor cells. Here we show that engagement of CD44 promotes CD44 cleavage and tumor cell migration, both of which were suppressed by a metalloproteinase inhibitor KB-R7785 and tissue inhibitor of metalloproteinases-1 (TIMP-1) but not by TIMP-2. We also present evidence that blockade of metalloproteinase-disintegrin ADAM10 (a disintegrin and metalloproteinase 10) by RNA interference suppresses CD44 cleavage induced by its ligation. Engagement of CD44 concurrently induced activation of the small GTPase Rac1 and led to drastic changes in cell morphology and actin cytoskeleton with redistribution of CD44 to newly generated membrane ruffling areas. A fluorescence resonance energy transfer approach to visualize GTP-bound Rac1 in living cells revealed the localization of the active Rac1 in the leading edge of the membrane ruffling areas upon ligation of CD44. Taken together, our results indicate that the cleavage of CD44 catalyzed by ADAM10 is augmented by the intracellular signaling elicited by engagement of CD44, through Rac-mediated cytoskeletal rearrangement, and suggest that CD44 cleavage contributes to the migration and invasion of tumor cells.
Collapse
Affiliation(s)
- Toshiyuki Murai
- Laboratory of Molecular and Cellular Recognition, Osaka University Graduate School of Medicine, Suita 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Thorne RF, Legg JW, Isacke CM. The role of the CD44 transmembrane and cytoplasmic domains in co-ordinating adhesive and signalling events. J Cell Sci 2004; 117:373-80. [PMID: 14702383 DOI: 10.1242/jcs.00954] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD44 is a widely distributed type I transmembrane glycoprotein and functions as the major hyaluronan receptor on most cell types. Although alternative splicing can produce a large number of different isoforms, they all retain the hyaluronan-binding Link-homology region and a common transmembrane and cytoplasmic domain, which are highly conserved between species. The past decade has seen an extensive investigation of this receptor owing to its importance in mediating cell-cell and cell-matrix interactions in both normal and disease states. Although roles for alternative splicing and variable glycosylation in determining ligand-binding interactions are now well established, the mechanisms by which CD44 integrates structural and signalling events to elicit cellular responses have been less well understood. However, there is now increasing evidence that CD44 is assembled in a regulated manner into membrane-cytoskeletal junctional complexes and, through both direct and indirect interactions, serves to focus downstream signal transduction events.
Collapse
Affiliation(s)
- Rick F Thorne
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | | | | |
Collapse
|
40
|
Park PW, Foster TJ, Nishi E, Duncan SJ, Klagsbrun M, Chen Y. Activation of syndecan-1 ectodomain shedding by Staphylococcus aureus alpha-toxin and beta-toxin. J Biol Chem 2003; 279:251-8. [PMID: 14573623 DOI: 10.1074/jbc.m308537200] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Exploitation of host components by microbes to promote their survival in the hostile host environment has been a recurring theme in recent years. Available data indicate that bacterial pathogens activate ectodomain shedding of host cell surface molecules to enhance their virulence. We reported previously that several major bacterial pathogens activate ectodomain shedding of syndecan-1, the major heparan sulfate proteoglycan of epithelial cells. Here we define the molecular basis of how Staphylococcus aureus activates syndecan-1 shedding. We screened mutant S. aureus strains devoid of various toxin and protease genes and found that only strains lacking both alpha-toxin and beta-toxin genes do not stimulate shedding. Mutations in the agr global regulatory locus, which positively regulates expression of alpha- and beta-toxins and other exoproteins, also abrogated the capacity to stimulate syndecan-1 shedding. Furthermore, purified S. aureus alpha- and beta-toxins, but not enterotoxin A and toxic shock syndrome toxin-1, rapidly potentiated shedding in a concentration-dependent manner. These results establish that S. aureus activates syndecan-1 ectodomain shedding via its two virulence factors, alpha- and beta-toxins. Toxin-activated shedding was also selectively inhibited by antagonists of the host cell shedding mechanism, indicating that alpha- and beta-toxins shed syndecan-1 ectodomains through stimulation of the host cell's shedding machinery. Interestingly, beta-toxin, but not alpha-toxin, also enhanced ectodomain shedding of syndecan-4 and heparin-binding epidermal growth factor. Because shedding of these ectodomains has been implicated in promoting bacterial pathogenesis, activation of ectodomain shedding by alpha-toxin and beta-toxin may be a previously unknown virulence mechanism of S. aureus.
Collapse
Affiliation(s)
- Pyong Woo Park
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Sugahara KN, Murai T, Nishinakamura H, Kawashima H, Saya H, Miyasaka M. Hyaluronan oligosaccharides induce CD44 cleavage and promote cell migration in CD44-expressing tumor cells. J Biol Chem 2003; 278:32259-65. [PMID: 12801931 DOI: 10.1074/jbc.m300347200] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CD44 is an adhesion molecule that serves as a cell surface receptor for several extracellular matrix components, including hyaluronan (HA). The proteolytic cleavage of CD44 from the cell surface plays a critical role in the migration of tumor cells. Although this cleavage can be induced by certain stimuli such as phorbol ester and anti-CD44 antibodies in vitro, the physiological inducer of CD44 cleavage in vivo is unknown. Here, we demonstrate that HA oligosaccharides of a specific size range induce CD44 cleavage from tumor cells. Fragmented HA containing 6-mers to 14-mers enhanced CD44 cleavage dose-dependently by interacting with CD44, whereas a large polymer HA failed to enhance CD44 cleavage, although it bound to CD44. Examination using uniformly sized HA oligosaccharides revealed that HAs smaller than 36 kDa significantly enhanced CD44 cleavage. In particular, the 6.9-kDa HA (36-mers) not only enhanced CD44 cleavage but also promoted tumor cell motility, which was completely inhibited by an anti-CD44 monoclonal antibody. These results raise the possibility that small HA oligosaccharides, which are known to occur in various tumor tissues, promote tumor invasion by enhancing the tumor cell motility that may be driven by CD44 cleavage.
Collapse
Affiliation(s)
- Kazuki N Sugahara
- Laboratory of Molecular and Cellular Recognition, Osaka University Graduate School of Medicine, 2-2, Yamada-oka, Suita 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
CD44 was once thought to simply be a transmembrane adhesion molecule that also played a role in the metabolism of its principal ligand hyaluronan. Investigations of CD44 over the past approximately 20 yr have established additional functions for CD44, including its capacity to mediate inflammatory cell function and tumor growth and metastasis. It has also become evident that intricate posttranslational modifications of CD44 regulate the affinity of the receptor for its ligands. In this review, we focus on emerging evidence that functional fragments of the cytoplasmic and ectodomain of CD44 can be liberated by enzymatic modification of cell surfaces as well as of cell-associated matrix. Based on the evidence discussed, we propose that CD44 exists in three phases, as a transmembrane receptor, as an integral component of the matrix, and as a soluble protein found in body fluids, each with biologically significant functions of which some are shared and some distinct. Thus, CD44 represents a model for understanding posttranslational processing and its emerging role as a general mechanism for regulating cell behavior.
Collapse
Affiliation(s)
- Joanna Cichy
- Jagiellonian University, Faculty of Biotechnology, 30-387 Kraków, Poland.
| | | |
Collapse
|
43
|
Boraldi F, Croce MA, Quaglino D, Sammarco R, Carnevali E, Tiozzo R, Pasquali-Ronchetti I. Cell-matrix interactions of in vitro human skin fibroblasts upon addition of hyaluronan. Tissue Cell 2003; 35:37-45. [PMID: 12589728 DOI: 10.1016/s0040-8166(02)00101-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Normal human skin fibroblasts were grown in a three-dimensional collagen gel or in monolayer in the presence or absence of high molecular weight hyaluronan (HA) to assess the influence of extracellular HA on cell-matrix interactions. HA incorporated into the collagen gel or added to the culture medium did not modify lattice retraction with time. The effect was independent from HA molecular weight (from 7.5 x 10(5) to 2.7 x 10(6) Da) and concentration (from 0.1 up to 1 mg/ml). HA did not affect shape and distribution of fibroblasts within the gel, whereas it induced the actin filaments to organise into thicker cables running underneath the plasma membrane. The same phenomenon was observed in fibroblasts grown in monolayer. By contrast, vimentin cytoskeleton and cell-substrate focal adhesions were not modified by exogenous HA. The number of fibroblasts attached to HA-coated dishes was always significantly lower compared to plastic and to collagen type I-coated plates. By contrast, adhesion was not affected by soluble HA added to the medium nor by anti-CD44 and anti-RHAMM-IHABP polyclonals. After 24-h seeding on collagen type I or on plastic, cells were large and spread. Conversely, cells adherent to HA-coated surfaces were long, thin and aligned into rows; alcian blue showed that cells were attached to the plastic in between HA bundles. Therefore, normal human skin fibroblasts exhibit very scarce, if any, adhesion to matrix HA, either soluble or immobilised. Moreover, even at high concentration, HA molecules do not exert any visco-mechanical effect on lattice retraction and do not interfere with fibroblast-collagen interactions nor with focal adhesion contacts of fibroblasts with the substrate. This is probably relevant in organogenesis and wound repair. By contrast, HA greatly modifies the organisation of the actin cytoskeleton, suggesting that CD44-mediated signal transduction by HA may affect cell locomotion and orientation, as indicated by the fusiform shape of fibroblasts grown in the presence of immobilised HA. A role of HA in cell orientation could be relevant for the deposition of collagen fibrils in regeneration and tissue remodelling.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Via Campi, 287, Modena 41100, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
CD44 is a multistructural and multifunctional cell surface molecule involved in cell proliferation, cell differentiation, cell migration, angiogenesis, presentation of cytokines, chemokines, and growth factors to the corresponding receptors, and docking of proteases at the cell membrane, as well as in signaling for cell survival. All these biological properties are essential to the physiological activities of normal cells, but they are also associated with the pathologic activities of cancer cells. Experiments in animals have shown that targeting of CD44 by antibodies, antisense,and CD44-soluble proteins markedly reduces the malignant activities of various neoplasms, stressing the therapeutic potential of anti-CD44 agents. Furthermore, because alternative splicing and posttranslational modifications generate many different CD44 sequences, including, perhaps, tumor-specific sequences, the production of anti-CD44 tumor-specific agents may be a realistic therapeutic approach. However, in many cancers (renal cancer and non-Hodgkin's lymphomas are exceptions), a high level of CD44 expression is not always associated with an unfavorable outcome. On the contrary, in some neoplams CD44 upregulation is associated with a favorable outcome. Even worse, in many cases different research grows analyzing the same neoplastic disease reached contradictory conclusions regarding the correlation between CD44 expression and disease prognosis, possibly due to differences in methodology. These problems must be resolved before applying anti-CD44 therapy to human cancers.
Collapse
Affiliation(s)
- David Naor
- The Lautenberg Center for General and Tumor Immunology, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | | | | | | | | |
Collapse
|
45
|
Mori H, Tomari T, Koshikawa N, Kajita M, Itoh Y, Sato H, Tojo H, Yana I, Seiki M. CD44 directs membrane-type 1 matrix metalloproteinase to lamellipodia by associating with its hemopexin-like domain. EMBO J 2002; 21:3949-59. [PMID: 12145196 PMCID: PMC126155 DOI: 10.1093/emboj/cdf411] [Citation(s) in RCA: 252] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Membrane-type 1 matrix metalloproteinase (MT1- MMP) localizes at the front of migrating cells and degrades the extracellular matrix barrier during cancer invasion. However, it is poorly understood how the polarized distribution of MT1-MMP at the migration front is regulated. Here, we demonstrate that MT1-MMP forms a complex with CD44H via the hemopexin-like (PEX) domain. A mutant MT1-MMP lacking the PEX domain failed to bind CD44H and did not localize at the lamellipodia. The cytoplasmic tail of CD44H, which comprises interfaces that associate with the actin cytoskeleton, was important for its localization at lamellipodia. Overexpression of a CD44H mutant lacking the cytoplasmic tail also prevented MT1-MMP from localizing at the lamellipodia. Modulation of F-actin with cytochalasin D revealed that both CD44H and MT1-MMP co-localize closely with the actin cytoskeleton, dependent on the cytoplasmic tail of CD44H. Thus, CD44H appears to act as a linker that connects MT1-MMP to the actin cytoskeleton and to play a role in directing MT1-MMP to the migration front. The PEX domain of MT1-MMP was indispensable in promoting cell migration and CD44H shedding.
Collapse
Affiliation(s)
| | | | | | | | - Yoshifumi Itoh
- Department of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Department of Molecular Virology and Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Graduate School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo, Japan and Department of Matrix Biology, Kennedy Institute of Rheumatology, Imperial College, Faculty of Medicine, Hammersmith, London, UK Corresponding author e-mail:
| | - Hiroshi Sato
- Department of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Department of Molecular Virology and Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Graduate School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo, Japan and Department of Matrix Biology, Kennedy Institute of Rheumatology, Imperial College, Faculty of Medicine, Hammersmith, London, UK Corresponding author e-mail:
| | - Hideaki Tojo
- Department of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Department of Molecular Virology and Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Graduate School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo, Japan and Department of Matrix Biology, Kennedy Institute of Rheumatology, Imperial College, Faculty of Medicine, Hammersmith, London, UK Corresponding author e-mail:
| | | | - Motoharu Seiki
- Department of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Department of Molecular Virology and Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Graduate School of Agricultural and Life Sciences, University of Tokyo, Bunkyo-ku, Tokyo, Japan and Department of Matrix Biology, Kennedy Institute of Rheumatology, Imperial College, Faculty of Medicine, Hammersmith, London, UK Corresponding author e-mail:
| |
Collapse
|
46
|
Hudrisier D, Bongrand P. Intercellular transfer of antigen-presenting cell determinants onto T cells: molecular mechanisms and biological significance. FASEB J 2002; 16:477-86. [PMID: 11919150 DOI: 10.1096/fj.01-0933rev] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Upon physiological stimulation, receptors with tyrosine kinase activity (RTK) are rapidly internalized together with their soluble ligands. T cell activation is the consequence of recognition by the T cell receptor (TCR) of specific peptide-major histocompatibility protein complexes (peptide-MHC) present at the membrane of antigen-presenting cells (APC). The TCR belongs to the RTK family and is known to be endocytosed upon ligand recognition. It differs from most other RTK in that its ligand, the peptide-MHC complex, is membrane bound and the TCR-ligand interaction is quite weak. Recent experiments have shown that the TCR ligand becomes internalized by T cells upon stimulation. Here we review current knowledge on the molecular mechanisms by which the membrane-bound MHC molecules can be transferred onto T cells, and propose hypotheses on the role this phenomenon could play in physio-pathological situations involving T cells.
Collapse
Affiliation(s)
- Denis Hudrisier
- INSERM U 395, CHU Purpan and Paul Sabatier University, BP3028 31024 Toulouse Cedex 3, France.
| | | |
Collapse
|