1
|
Takeda M, Theardy MS, Sorokin A, Coker O, Kanikarla P, Chen S, Yang Z, Nguyen P, Wei Y, Yao J, Wang X, Yan L, Jin Y, Cai Y, Paku M, Chen Z, Li KZ, Citron F, Tomihara H, Gao S, Deem AK, Zhao J, Wang H, Hanash S, DePinho RA, Maitra A, Draetta GF, Ying H, Kopetz S, Yao W. Therapeutic targeting of Syndecan-1 axis overcomes acquired resistance to KRAS-targeted therapy in gastrointestinal cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606865. [PMID: 39211217 PMCID: PMC11361106 DOI: 10.1101/2024.08.06.606865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The therapeutic benefit of recently developed mutant KRAS (mKRAS) inhibitors has been limited by the rapid onset of resistance. Here, we aimed to delineate the mechanisms underlying acquired resistance to mKRAS inhibition and identify actionable targets for overcoming this clinical challenge. Previously, we identified Syndecan-1 (SDC1) as a key effector for pancreatic cancer progression whose surface expression is driven by mKRAS. By leveraging both pancreatic and colorectal cancer models, we found that surface SDC1 expression was initially diminished upon mKRAS inhibition, but recovered in tumor cells that bypass mKRAS dependency. Functional studies showed that these tumors depended on SDC1 for survival, further establishing SDC1 as a driver for the acquired resistance to mKRAS inhibition. Mechanistically, we revealed that the YAP1-SDC1 axis was the major driving force for bypassing mKRAS dependency to sustain nutrient salvage machinery and tumor maintenance. Specifically, YAP1 activation mediated the recovery of SDC1 localization on cell surface that sustained macropinocytosis and enhanced the activation of multiple RTKs, promoting resistance to KRAS-targeted therapy. Overall, our study has provided the rationale for targeting the YAP-SDC1 axis to overcome resistance to mKRAS inhibition, thereby revealing new therapeutic opportunities for improving the clinical outcome of patients with KRAS-mutated cancers.
Collapse
|
2
|
Single cell multi-omic reference atlases of non-human primate immune tissues reveals CD102 as a biomarker for long-lived plasma cells. Commun Biol 2022; 5:1399. [PMID: 36543997 PMCID: PMC9770566 DOI: 10.1038/s42003-022-04216-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/02/2022] [Indexed: 12/24/2022] Open
Abstract
In response to infection or immunization, antibodies are produced that provide protection against re-exposure with the same pathogen. These antibodies can persist at high titers for decades and are maintained by bone marrow-resident long-lived plasma cells (LLPC). However, the durability of antibody responses to immunization varies amongst vaccines. It is unknown what factors contribute to the differential longevity of serum antibody responses and whether heterogeneity in LLPC contributes to this phenomenon. While LLPC differentiation has been studied extensively in mice, little is known about this population in humans or non-human primates (NHP). Here, we use multi-omic single-cell profiling to identify and characterize the LLPC compartment in NHP. We identify LLPC biomarkers including the marker CD102 and show that CD102 in combination with CD31 identifies LLPC in NHP bone marrow. Additionally, we find that CD102 is expressed by LLPC in mouse and humans. These results further our understanding of the LLPC compartment in NHP, identify biomarkers of LLPC, and provide tissue-specific single cell references for future studies.
Collapse
|
3
|
The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev 2022; 181:114069. [PMID: 34838648 PMCID: PMC8860232 DOI: 10.1016/j.addr.2021.114069] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Comprehensive overview of different classes of ECM molecules in the HSC niche. Overview of current knowledge on role of biophysics of the HSC niche. Description of approaches to create artificial stem cell niches for several application. Importance of considering ECM in drug development and testing.
Hematopoietic stem cells (HSCs) are the life-long source of all types of blood cells. Their function is controlled by their direct microenvironment, the HSC niche in the bone marrow. Although the importance of the extracellular matrix (ECM) in the niche by orchestrating niche architecture and cellular function is widely acknowledged, it is still underexplored. In this review, we provide a comprehensive overview of the ECM in HSC niches. For this purpose, we first briefly outline HSC niche biology and then review the role of the different classes of ECM molecules in the niche one by one and how they are perceived by cells. Matrix remodeling and the emerging importance of biophysics in HSC niche function are discussed. Finally, the application of the current knowledge of ECM in the niche in form of artificial HSC niches for HSC expansion or targeted differentiation as well as drug testing is reviewed.
Collapse
|
4
|
Klein K, Hölzemer A, Wang T, Kim TE, Dugan HL, Jost S, Altfeld M, Garcia-Beltran WF. A Genome-Wide CRISPR/Cas9-Based Screen Identifies Heparan Sulfate Proteoglycans as Ligands of Killer-Cell Immunoglobulin-Like Receptors. Front Immunol 2021; 12:798235. [PMID: 34917099 PMCID: PMC8669139 DOI: 10.3389/fimmu.2021.798235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
While human leukocyte antigen (HLA) and HLA-like proteins comprise an overwhelming majority of known ligands for NK-cell receptors, the interactions of NK-cell receptors with non-conventional ligands, particularly carbohydrate antigens, is less well described. We previously found through a bead-based HLA screen that KIR3DS1, a formerly orphan member of the killer-cell immunoglobulin-like receptor (KIR) family, binds to HLA-F. In this study, we assessed the ligand binding profile of KIR3DS1 to cell lines using Fc fusion constructs, and discovered that KIR3DS1-Fc exhibited binding to several human cell lines including ones devoid of HLA. To identify these non-HLA ligands, we developed a magnetic enrichment-based genome-wide CRISPR/Cas9 knock-out screen approach, and identified enzymes involved in the biosynthesis of heparan sulfate as crucial for the binding of KIR3DS1-Fc to K562 cells. This interaction between KIR3DS1 and heparan sulfate was confirmed via surface plasmon resonance, and removal of heparan sulfate proteoglycans from cell surfaces abolished KIR3DS1-Fc binding. Testing of additional KIR-Fc constructs demonstrated that KIR family members containing a D0 domain (KIR3DS1, KIR3DL1, KIR3DL2, KIR2DL4, and KIR2DL5) bound to heparan sulfate, while those without a D0 domain (KIR2DL1, KIR2DL2, KIR2DL3, and KIR2DS4) did not. Overall, this study demonstrates the use of a genome-wide CRISPR/Cas9 knock-out strategy to unbiasedly identify unconventional ligands of NK-cell receptors. Furthermore, we uncover a previously underrecognized binding of various activating and inhibitory KIRs to heparan sulfate proteoglycans that may play a role in NK-cell receptor signaling and target-cell recognition.
Collapse
Affiliation(s)
- Klara Klein
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Angelique Hölzemer
- Leibniz Institute for Experimental Virology, Hamburg, Germany
- First Department of Internal Medicine, Division of Infectious Diseases, University Medical Centre Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Tim Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Whitehead Institute for Biomedical Research, Cambridge, MA, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Tae-Eun Kim
- Ragon Institute of Massachusetts General Hospital (MGH), MIT, and Harvard, Cambridge, MA, United States
| | - Haley L. Dugan
- Ragon Institute of Massachusetts General Hospital (MGH), MIT, and Harvard, Cambridge, MA, United States
- Adimab, LLC, Lebanon, NH, United States
| | - Stephanie Jost
- Ragon Institute of Massachusetts General Hospital (MGH), MIT, and Harvard, Cambridge, MA, United States
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Marcus Altfeld
- Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Wilfredo F. Garcia-Beltran
- Ragon Institute of Massachusetts General Hospital (MGH), MIT, and Harvard, Cambridge, MA, United States
- Department of Pathology, Massachusetts General Hospital (MGH), Boston, MA, United States
- *Correspondence: Wilfredo F. Garcia-Beltran,
| |
Collapse
|
5
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Ross-Munro E, Kwa F, Kreiner J, Khore M, Miller SL, Tolcos M, Fleiss B, Walker DW. Midkine: The Who, What, Where, and When of a Promising Neurotrophic Therapy for Perinatal Brain Injury. Front Neurol 2020; 11:568814. [PMID: 33193008 PMCID: PMC7642484 DOI: 10.3389/fneur.2020.568814] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Midkine (MK) is a small secreted heparin-binding protein highly expressed during embryonic/fetal development which, through interactions with multiple cell surface receptors promotes growth through effects on cell proliferation, migration, and differentiation. MK is upregulated in the adult central nervous system (CNS) after multiple types of experimental injury and has neuroprotective and neuroregenerative properties. The potential for MK as a therapy for developmental brain injury is largely unknown. This review discusses what is known of MK's expression and actions in the developing brain, areas for future research, and the potential for using MK as a therapeutic agent to ameliorate the effects of brain damage caused by insults such as birth-related hypoxia and inflammation.
Collapse
Affiliation(s)
- Emily Ross-Munro
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Faith Kwa
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Jenny Kreiner
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Madhavi Khore
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| | - Mary Tolcos
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Bobbi Fleiss
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,Neurodiderot, Inserm U1141, Universita de Paris, Paris, France
| | - David W Walker
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| |
Collapse
|
7
|
Teixeira FCOB, Götte M. Involvement of Syndecan-1 and Heparanase in Cancer and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:97-135. [PMID: 32274708 DOI: 10.1007/978-3-030-34521-1_4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cell surface heparan sulfate proteoglycan Syndecan-1 acts as an important co-receptor for receptor tyrosine kinases and chemokine receptors, and as an adhesion receptor for structural glycoproteins of the extracellular matrix. It serves as a substrate for heparanase, an endo-β-glucuronidase that degrades specific domains of heparan sulfate carbohydrate chains and thereby alters the functional status of the proteoglycan and of Syndecan-1-bound ligands. Syndecan-1 and heparanase show multiple levels of functional interactions, resulting in mutual regulation of their expression, processing, and activity. These interactions are of particular relevance in the context of inflammation and malignant disease. Studies in animal models have revealed a mechanistic role of Syndecan-1 and heparanase in the regulation of contact allergies, kidney inflammation, multiple sclerosis, inflammatory bowel disease, and inflammation-associated tumorigenesis. Moreover, functional interactions between Syndecan-1 and heparanase modulate virtually all steps of tumor progression as defined in the Hallmarks of Cancer. Due to their prognostic value in cancer, and their mechanistic involvement in tumor progression, Syndecan-1 and heparanase have emerged as important drug targets. Data in preclinical models and preclinical phase I/II studies have already yielded promising results that provide a translational perspective.
Collapse
Affiliation(s)
- Felipe C O B Teixeira
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.
| |
Collapse
|
8
|
Zhao B, Xu Y, Zhao Y, Shen S, Sun Q. Identification of Potential Key Genes Associated With the Pathogenesis, Metastasis, and Prognosis of Triple-Negative Breast Cancer on the Basis of Integrated Bioinformatics Analysis. Front Oncol 2020; 10:856. [PMID: 32596149 PMCID: PMC7304260 DOI: 10.3389/fonc.2020.00856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
Objective: Breast cancer is the most common solid tumor affecting women and the second leading cause of cancer-related death worldwide, and triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer. We aimed to identify potential TNBC-specific therapeutic targets by performing an integrative analysis on previously published TNBC transcriptome microarray data. Methods: Differentially expressed genes (DEGs) between TNBC and normal breast tissues were screened using six Gene Expression Omnibus (GEO) datasets, and DEGs between metastatic TNBC and non-metastatic TNBC were screened using one GEO dataset. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses were performed on the overlapping DEGs. The Cancer Genome Atlas (TCGA) TNBC data were used to identify candidate genes that were strongly associated with survival. Expression of the candidate genes in TNBC cell lines was blocked or augmented using a lentivirus system, and transwell assays were used to determine their effect on TNBC migration. Results: Eight upregulated genes and nine downregulated genes were found to be differentially expressed both between TNBC and normal breast tissues and between metastatic TNBC and non-metastatic TNBC. Among them, S100P and SDC1 were identified as poor prognostic genes. Furthermore, compared with control cells, SDC1-overexpressing TNBC cells showed enhanced migration ability, whereas SDC1 knockdown markedly reduced the migration of TNBC cells. Conclusion: Our study determined that S100P and SDC1 may be potential treatment targets as well as prognostic biomarkers of TNBC.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yali Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yang Zhao
- Department of Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
9
|
The endothelial glycocalyx anchors von Willebrand factor fibers to the vascular endothelium. Blood Adv 2019; 2:2347-2357. [PMID: 30237293 DOI: 10.1182/bloodadvances.2017013995] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 08/03/2018] [Indexed: 12/22/2022] Open
Abstract
The dynamic change from a globular conformation to an elongated fiber determines the ability of von Willebrand factor (VWF) to trap platelets. Fiber formation is favored by the anchorage of VWF to the endothelial cell surface, and VWF-platelet aggregates on the endothelium contribute to inflammation, infection, and tumor progression. Although P-selectin and ανβ3-integrins may bind VWF, their precise role is unclear, and additional binding partners have been proposed. In the present study, we evaluated whether the endothelial glycocalyx anchors VWF fibers to the endothelium. Using microfluidic experiments, we showed that stabilization of the endothelial glycocalyx by chitosan oligosaccharides or overexpression of syndecan-1 (SDC-1) significantly supports the binding of VWF fibers to endothelial cells. Heparinase-mediated degradation or impaired synthesis of heparan sulfate (HS), a major component of the endothelial glycocalyx, reduces VWF fiber-dependent platelet recruitment. Molecular interaction studies using flow cytometry and live-cell fluorescence microscopy provided further evidence that VWF binds to HS linked to SDC-1. In a murine melanoma model, we found that protection of the endothelial glycocalyx through the silencing of heparanase increases the number of VWF fibers attached to the wall of tumor blood vessels. In conclusion, we identified HS chains as a relevant binding factor for VWF fibers at the endothelial cell surface in vitro and in vivo.
Collapse
|
10
|
Role of cell surface proteoglycans in cancer immunotherapy. Semin Cancer Biol 2019; 62:48-67. [PMID: 31336150 DOI: 10.1016/j.semcancer.2019.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/23/2022]
Abstract
Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.
Collapse
|
11
|
Gougoula C, Bielfeld AP, Pour SJ, Sager M, Krüssel JS, Benten WPM, Baston-Büst DM. Metabolic and behavioral parameters of mice with reduced expression of Syndecan-1. PLoS One 2019; 14:e0219604. [PMID: 31299063 PMCID: PMC6625734 DOI: 10.1371/journal.pone.0219604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/27/2019] [Indexed: 11/19/2022] Open
Abstract
Energy balance is essential for all species. Ligand-receptor interactions mediate processes that regulate body activities like reproduction and metabolism based on the energy status. Such receptors are the heparan sulfate proteoglycans and specifically the family of syndecans. Therefore we investigated the differences of metabolic parameters of heterozygous Syndecan 1 mice (Sdc1+/-) with reduced expression of Sdc1 and the corresponding wild type mice. Sdc1+/- mice have a reduced body weight although they show increased leptin and decreased corticosterone levels. Furthermore, their food and water intake is increased. This is accompanied with less adipose tissue, smaller adipocytes and thus an increased density of adipocytes. For the detailed analysis of the metabolism the automated PhenoMaster system has been used, which allowed continuous and undisturbed recording of food and water intake, energy expenditure and movement. The reason for the lower body weight was the higher energy expenditure of these animals compared to controls. Additionally, female Sdc1+/- mice showed an increased locomotor activity. Referring to organs, the intestine in Sdc1+/- mice was heavier and longer, but no differences at the cellular level could be observed. These findings were independent of normal mating or vice versa embryo transfers of Sdc1+/- and wild type embryos in recipient females of the other genotype. Herein we showed that the reduced expression of Sdc1 led to an altered metabolism on fetal as well as on maternal side, which may play a role in the growth restriction observed in human pregnancy pathologies and in mice lacking Sdc1.
Collapse
Affiliation(s)
- Christina Gougoula
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT) of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | | | - Sarah Jean Pour
- Düsseldorf University Hospital, Department of OB/GYN and REI (UniKiD), Düsseldorf, Germany
| | - Martin Sager
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT) of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - Jan-Steffen Krüssel
- Düsseldorf University Hospital, Department of OB/GYN and REI (UniKiD), Düsseldorf, Germany
| | - Wilhelm Peter M. Benten
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT) of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - Dunja Maria Baston-Büst
- Düsseldorf University Hospital, Department of OB/GYN and REI (UniKiD), Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
12
|
Kasza I, Adler D, Nelson DW, Eric Yen CL, Dumas S, Ntambi JM, MacDougald OA, Hernando D, Porter WP, Best FA, Alexander CM. Evaporative cooling provides a major metabolic energy sink. Mol Metab 2019; 27:47-61. [PMID: 31302039 PMCID: PMC6717770 DOI: 10.1016/j.molmet.2019.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/08/2019] [Accepted: 06/27/2019] [Indexed: 11/30/2022] Open
Abstract
Objective Elimination of food calories as heat could help redress the excess accumulation of metabolic energy exhibited as obesity. Prior studies have focused on the induction of thermogenesis in beige and brown adipose tissues as the application of this principle, particularly because the β-adrenergic environment associated with thermogenic activation has been shown to have positive health implications. The counterpoint to this strategy is the regulation of heat loss; we propose that mammals with inefficient heat conservation will require more thermogenesis to maintain body temperature. Methods Surface temperature thermography and rates of trans-epidermal water loss were integrated to profile the total heat transfer of genetically-engineered and genetically variable mice. Results These data were incorporated with energy expenditure data to generate a biophysical profile to test the significance of increased rates of evaporative cooling. Conclusions We show that mouse skins vary considerably in their heat retention properties, whether because of naturally occurring variation (SKH-1 mice), or genetic modification of the heat-retaining lipid lamellae (SCD1, DGAT1 or Agouti Ay obese mice). In particular, we turn attention to widely different rates of evaporative cooling as the result of trans-epidermal water loss; higher rates of heat loss by evaporative cooling leads to increased demand for thermogenesis. We speculate that this physiology could be harnessed to create an energy sink to assist with strategies aimed at treating metabolic diseases.
Collapse
Affiliation(s)
- Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, United States
| | - Doug Adler
- Space Science and Engineering Center, University of Wisconsin-Madison, United States
| | - David W Nelson
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - C-L Eric Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - Sabrina Dumas
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States
| | - James M Ntambi
- Department of Nutritional Sciences, University of Wisconsin-Madison, United States; Department of Biochemistry, University of Wisconsin-Madison, United States
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan, United States
| | - Diego Hernando
- Department of Radiology, University of Wisconsin-Madison, United States
| | - Warren P Porter
- Department of Zoology, University of Wisconsin-Madison, United States
| | - Fred A Best
- Space Science and Engineering Center, University of Wisconsin-Madison, United States
| | - C M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, United States.
| |
Collapse
|
13
|
Vitale D, Kumar Katakam S, Greve B, Jang B, Oh ES, Alaniz L, Götte M. Proteoglycans and glycosaminoglycans as regulators of cancer stem cell function and therapeutic resistance. FEBS J 2019; 286:2870-2882. [PMID: 31230410 DOI: 10.1111/febs.14967] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022]
Abstract
In contrast to the bulk of the tumor, a subset of cancer cells called cancer stem cells (CSC; or tumor-initiating cells) is characterized by self-renewal, unlimited proliferative potential, expression of multidrug resistance proteins, active DNA repair capacity, apoptosis resistance, and a considerable developmental plasticity. Due to these properties, CSCs display increased resistance to chemo- and radiotherapy. Recent findings indicate that aberrant functions of proteoglycans (PGs) and glycosaminoglycans (GAGs) contribute substantially to the CSC phenotype and therapeutic resistance. In this review, we summarize how the diverse functions of the glycoproteins and carbohydrates facilitate acquisition and maintenance of the CSC phenotype, and how this knowledge can be exploited to develop novel anticancer therapies. For example, the large transmembrane chondroitin sulfate PG NG2/CSPG4 marks stem cell (SC) populations in brain tumors. Cell surface heparan sulfate PGs of the syndecan and glypican families modulate the stemness-associated Wnt, hedgehog, and notch signaling pathways, whereas the interplay of hyaluronan in the SC niche with CSC CD44 determines the maintenance of stemness and promotes therapeutic resistance. A better understanding of the molecular mechanisms by which PGs and GAGs regulate CSC function will aid the development of targeted therapeutic approaches which could avoid relapse after an otherwise successful conventional therapy. Chimeric antigen receptor T cells, PG-primed dendritic cells, PG-targeted antibody-drug conjugates, and inhibitory peptides and glycans have already shown highly promising results in preclinical models.
Collapse
Affiliation(s)
- Daiana Vitale
- Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín, Argentina
| | | | - Burkhard Greve
- Department of Radiotherapy - Radiooncology, Münster University Hospital, Germany
| | - Bohee Jang
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Eok-Soo Oh
- Department of Life Sciences, The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Laura Alaniz
- Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín, Argentina
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Germany
| |
Collapse
|
14
|
Gruszka AM, Valli D, Restelli C, Alcalay M. Adhesion Deregulation in Acute Myeloid Leukaemia. Cells 2019; 8:E66. [PMID: 30658474 PMCID: PMC6356639 DOI: 10.3390/cells8010066] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion is a process through which cells interact with and attach to neighboring cells or matrix using specialized surface cell adhesion molecules (AMs). Adhesion plays an important role in normal haematopoiesis and in acute myeloid leukaemia (AML). AML blasts express many of the AMs identified on normal haematopoietic precursors. Differential expression of AMs between normal haematopoietic cells and leukaemic blasts has been documented to a variable extent, likely reflecting the heterogeneity of the disease. AMs govern a variety of processes within the bone marrow (BM), such as migration, homing, and quiescence. AML blasts home to the BM, as the AM-mediated interaction with the niche protects them from chemotherapeutic agents. On the contrary, they detach from the niches and move from the BM into the peripheral blood to colonize other sites, i.e., the spleen and liver, possibly in a process that is reminiscent of epithelial-to-mesenchymal-transition in metastatic solid cancers. The expression of AMs has a prognostic impact and there are ongoing efforts to therapeutically target adhesion in the fight against leukaemia.
Collapse
Affiliation(s)
- Alicja M Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Cecilia Restelli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy.
| |
Collapse
|
15
|
Chute C, Yang X, Meyer K, Yang N, O'Neil K, Kasza I, Eliceiri K, Alexander C, Friedl A. Syndecan-1 induction in lung microenvironment supports the establishment of breast tumor metastases. Breast Cancer Res 2018; 20:66. [PMID: 29976229 PMCID: PMC6034333 DOI: 10.1186/s13058-018-0995-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/28/2018] [Indexed: 01/04/2023] Open
Abstract
Background Syndecan-1 (Sdc1), a cell surface heparan sulfate proteoglycan normally expressed primarily by epithelia and plasma cells, is aberrantly induced in stromal fibroblasts of breast carcinomas. Stromal fibroblast-derived Sdc1 participates in paracrine growth stimulation of breast carcinoma cells and orchestrates stromal extracellular matrix fiber alignment, thereby creating a migration and invasion-permissive microenvironment. Here, we specifically tested the role of stromal Sdc1 in metastasis. Methods The metastatic potential of the aggressive mouse mammary carcinoma cell lines, 4T1 and E0776, was tested in wild-type and genetically Sdc1-deficient host animals. Metastatic lesions were characterized by immunohistochemical analysis. Results After orthotopic inoculation, the lung metastatic burden was reduced in Sdc1−/− animals by 97% and more than 99%, in BALB/cJ and C57BL/6 animals, respectively. The difference in metastatic efficiency was maintained when the tumor cells were injected into the tail vein, suggesting that host Sdc1 exerts its effect during later stages of the metastatic cascade. Co-localization studies identified Sdc1 expression in stromal fibroblasts within the metastatic microenvironment and in normal airway epithelial cells but not in other cells (endothelial cells, α-smooth muscle actin positive cells, leucocytes, macrophages). The Ki67 proliferation index and the rate of apoptosis of the metastatic tumor cells were diminished in Sdc1−/− vs. Sdc1+/+ animals, and leucocyte density was indistinguishable. Sdc1-mediated metastatic efficiency was abolished when the animals were housed at a thermoneutral ambient temperature of 31 °C, suggesting that the host Sdc1 effect on metastasis requires mild cold stress. Conclusions In summary, Sdc1 is induced in the lung microenvironment after mammary carcinoma cell dissemination and promotes outgrowth of metastases in a temperature-dependent manner. Electronic supplementary material The online version of this article (10.1186/s13058-018-0995-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Colleen Chute
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 6051 WIMR, MC-2275, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Xinhai Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 6051 WIMR, MC-2275, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Kristy Meyer
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 6051 WIMR, MC-2275, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Ning Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 6051 WIMR, MC-2275, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Keelin O'Neil
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 6051 WIMR, MC-2275, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Ildiko Kasza
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Kevin Eliceiri
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI, USA.,Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline Alexander
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - Andreas Friedl
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 6051 WIMR, MC-2275, 1111 Highland Avenue, Madison, WI, 53705, USA. .,Pathology and Laboratory Medicine Service, William S. Middleton Memorial Veterans Hospital, Department of Veterans Affairs Medical Center, Madison, WI, USA. .,University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
16
|
Ionizing radiation-mediated premature senescence and paracrine interactions with cancer cells enhance the expression of syndecan 1 in human breast stromal fibroblasts: the role of TGF-β. Aging (Albany NY) 2017; 8:1650-69. [PMID: 27434331 PMCID: PMC5032688 DOI: 10.18632/aging.100989] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/26/2016] [Indexed: 12/12/2022]
Abstract
The cell surface proteoglycan syndecan 1 (SDC1) is overexpressed in the malignant breast stromal fibroblasts, creating a favorable milieu for tumor cell growth. In the present study, we found that ionizing radiation, a well-established treatment in human breast cancer, provokes premature senescence of human breast stromal fibroblasts in vitro, as well as in the breast tissue in vivo. These senescent cells were found to overexpress SDC1 both in vitro and in vivo. By using a series of specific inhibitors and siRNA approaches, we showed that this SDC1 overexpression in senescent cells is the result of an autocrine action of Transforming Growth Factor-β (TGF-β) through the Smad pathway and the transcription factor Sp1, while the classical senescence pathways of p53 or p38 MAPK - NF-kB are not involved. In addition, the highly invasive human breast cancer cells MDA-MB-231 (in contrast to the low-invasive MCF-7) can also enhance SDC1 expression, both in early-passage and senescent fibroblasts via a paracrine action of TGF-β. The above suggest that radiation-mediated premature senescence and invasive tumor cells, alone or in combination, enhance SDC1 expression in breast stromal fibroblasts, a poor prognostic factor for cancer growth, and that TGF-β plays a crucial role in this process.
Collapse
|
17
|
Pal-Ghosh S, Tadvalkar G, Stepp MA. Alterations in Corneal Sensory Nerves During Homeostasis, Aging, and After Injury in Mice Lacking the Heparan Sulfate Proteoglycan Syndecan-1. Invest Ophthalmol Vis Sci 2017; 58:4959-4975. [PMID: 28973369 PMCID: PMC5627677 DOI: 10.1167/iovs.17-21531] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Purpose To determine the impact of the loss of syndecan 1 (SDC1) on intraepithelial corneal nerves (ICNs) during homeostasis, aging, and in response to 1.5-mm trephine and debridement injury. Methods Whole-mount corneas are used to quantify ICN density and thickness over time after birth and in response to injury in SDC1-null and wild-type (WT) mice. High-resolution three-dimensional imaging is used to visualize intraepithelial nerve terminals (INTs), axon fragments, and lysosomes in corneal epithelial cells using antibodies against growth associated protein 43 (GAP43), βIII tubulin, and LAMP1. Quantitative PCR was performed to quantify expression of SDC1, SDC2, SDC3, and SDC4 in corneal epithelial mRNA. Phagocytosis was assessed by quantifying internalization of fluorescently labeled 1-μm latex beads. Results Intraepithelial corneal nerves innervate the corneas of SDC1-null mice more slowly. At 8 weeks, ICN density is less but thickness is greater. Apically projecting intraepithelial nerve terminals and lysosome-associated membrane glycoprotein 1 (LAMP1) are also reduced in unwounded SDC1-null corneas. Quantitative PCR and immunofluorescence studies show that SDC3 expression and localization are increased in SDC1-null ICNs. Wild-type and SDC1-null corneas lose ICN density and thickness as they age. Recovery of axon density and thickness after trephine but not debridement wounds is slower in SDC1-null corneas compared with WT. Experiments assessing phagocytosis show reduced bead internalization by SDC1-null epithelial cells. Conclusions Syndecan-1 deficiency alters ICN morphology and homeostasis during aging, reduces epithelial phagocytosis, and impairs reinnervation after trephine but not debridement injury. These data provide insight into the mechanisms used by sensory nerves to reinnervate after injury.
Collapse
Affiliation(s)
- Sonali Pal-Ghosh
- Department of Anatomy and Regenerative Biology, The George Washington University Medical School, Washington, D.C., United States
| | - Gauri Tadvalkar
- Department of Anatomy and Regenerative Biology, The George Washington University Medical School, Washington, D.C., United States
| | - Mary Ann Stepp
- Department of Anatomy and Regenerative Biology, The George Washington University Medical School, Washington, D.C., United States.,Department of Ophthalmology, The George Washington University Medical School, Washington, D.C., United States
| |
Collapse
|
18
|
Ibrahim SA, Gadalla R, El-Ghonaimy EA, Samir O, Mohamed HT, Hassan H, Greve B, El-Shinawi M, Mohamed MM, Götte M. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Mol Cancer 2017; 16:57. [PMID: 28270211 PMCID: PMC5341174 DOI: 10.1186/s12943-017-0621-z] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 02/22/2017] [Indexed: 12/15/2022] Open
Abstract
Background Inflammatory breast cancer (IBC), a particularly aggressive form of breast cancer, is characterized by cancer stem cell (CSC) phenotype. Due to a lack of targeted therapies, the identification of molecular markers of IBC is of major importance. The heparan sulfate proteoglycan Syndecan-1 acts as a coreceptor for growth factors and chemokines, modulating inflammation, tumor progression, and cancer stemness, thus it may emerge as a molecular marker for IBC. Methods We characterized expression of Syndecan-1 and the CSC marker CD44, Notch-1 & -3 and EGFR in carcinoma tissues of triple negative IBC (n = 13) and non-IBC (n = 17) patients using qPCR and immunohistochemistry. Impact of siRNA-mediated Syndecan-1 knockdown on the CSC phenotype of the human triple negative IBC cell line SUM-149 and HER-2-overexpressing non-IBC SKBR3 cells employing qPCR, flow cytometry, Western blotting, secretome profiling and Notch pharmacological inhibition experiments. Data were statistically analyzed using Student’s t-test/Mann-Whitney U-test or one-way ANOVA followed by Tukey’s multiple comparison tests. Results Our data indicate upregulation and a significant positive correlation of Syndecan-1 with CD44 protein, and Notch-1 & -3 and EGFR mRNA in IBC vs non-IBC. ALDH1 activity and the CD44(+)CD24(-/low) subset as readout of a CSC phenotype were reduced upon Syndecan-1 knockdown. Functionally, Syndecan-1 silencing significantly reduced 3D spheroid and colony formation. Intriguingly, qPCR results indicate downregulation of the IL-6, IL-8, CCL20, gp130 and EGFR mRNA upon Syndecan-1 suppression in both cell lines. Moreover, Syndecan-1 silencing significantly downregulated Notch-1, -3, -4 and Hey-1 in SUM-149 cells, and downregulated only Notch-3 and Gli-1 mRNA in SKBR3 cells. Secretome profiling unveiled reduced IL-6, IL-8, GRO-alpha and GRO a/b/g cytokines in conditioned media of Syndecan-1 knockdown SUM-149 cells compared to controls. The constitutively activated STAT3 and NFκB, and expression of gp130, Notch-1 & -2, and EGFR proteins were suppressed upon Syndecan-1 ablation. Mechanistically, gamma-secretase inhibition experiments suggested that Syndecan-1 may regulate the expression of IL-6, IL-8, gp130, Hey-1, EGFR and p-Akt via Notch signaling. Conclusions Syndecan-1 acts as a novel tissue biomarker and a modulator of CSC phenotype of triple negative IBC via the IL-6/STAT3, Notch and EGFR signaling pathways, thus emerging as a promising therapeutic target for IBC. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0621-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Ramy Gadalla
- Department of Zoology, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Eslam A El-Ghonaimy
- Department of Zoology, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Omnia Samir
- Department of Zoology, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Hossam Taha Mohamed
- Department of Zoology, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Hebatallah Hassan
- Department of Zoology, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, University Hospital Münster, Münster, Germany
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, 11566, Cairo, Egypt
| | | | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149, Münster, Germany.
| |
Collapse
|
19
|
Pasqualon T, Pruessmeyer J, Jankowski V, Babendreyer A, Groth E, Schumacher J, Koenen A, Weidenfeld S, Schwarz N, Denecke B, Jahr H, Dreymueller D, Jankowski J, Ludwig A. A cytoplasmic C-terminal fragment of Syndecan-1 is generated by sequential proteolysis and antagonizes Syndecan-1 dependent lung tumor cell migration. Oncotarget 2016; 6:31295-312. [PMID: 26378057 PMCID: PMC4741606 DOI: 10.18632/oncotarget.5174] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/20/2015] [Indexed: 12/29/2022] Open
Abstract
Syndecan-1 is a surface expressed heparan sulphate proteoglycan, which is upregulated by several tumor types and involved in tumor cell migration and metastasis. Syndecan-1 is shed from the cell surface and the remaining transmembrane fragment undergoes intramembrane proteolysis by γ-secretase. We here show that this generates a cytoplasmic C-terminal fragment (cCTF). In epithelial lung tumor A549 cells the endogenously produced cCTF accumulated when its proteasomal degradation was blocked with bortezomib and this accumulation was prevented by γ-secretase inhibition. Overexpression of the cCTF suppressed migration and invasion of A549 cells. This inhibitory effect was only seen when endogenous syndecan-1 was present, but not in syndecan-1 deficient cells. Further, overexpression of syndecan-1 cCTF increased the basal activation of Src kinase, focal adhesion kinase (FAK) and Rho GTPase. This was associated with increased adhesion to fibronectin and collagen G and an increased recruitment of paxillin to focal adhesions. Moreover, lung tumor formation of A549 cells in mice was reduced by overexpression of syndecan-1 cCTF. Finally, delivery of a synthetic peptide corresponding to the syndecan-1 cCTF suppressed A549 cell migration and increased basal phosphorylation of Src and FAK. Our data indicate that the syndecan-1 cCTF antagonizes syndecan-1 dependent tumor cell migration in vitro and in vivo by dysregulating proadhesive signaling pathways and suggest that the cCTF can be used as an inhibitory peptide.
Collapse
Affiliation(s)
- Tobias Pasqualon
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Jessica Pruessmeyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Vera Jankowski
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Esther Groth
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Julian Schumacher
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Andrea Koenen
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Sarah Weidenfeld
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Nicole Schwarz
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
| | - Holger Jahr
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
20
|
Kasza I, Hernando D, Roldán-Alzate A, Alexander CM, Reeder SB. Thermogenic profiling using magnetic resonance imaging of dermal and other adipose tissues. JCI Insight 2016; 1:e87146. [PMID: 27668285 DOI: 10.1172/jci.insight.87146] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dermal white adipose tissue (dWAT) was recently recognized for its potential to modify whole body metabolism. Here, we show that dWAT can be quantified using a high-resolution, fat-specific magnetic resonance imaging (MRI) technique. Noninvasive MRI has been used to describe adipocyte depots for many years; the MRI technique we describe uses an advanced fat-specific method to measure the thickness of dWAT, together with the total volume of WAT and the relative activation/fat depletion of brown adipose tissues (BAT). Since skin-embedded adipocytes may provide natural insulation, they provide an important counterpoint to the activation of thermogenic brown and beige adipose tissues, whereby these distinct depots are functionally interrelated and require simultaneous assay. This method was validated using characterized mouse cohorts of a lipodystrophic, dWAT-deficient strain (syndecan-1 KO) and 2 obese models (diet-induced obese mice and genetically obese animals, ob/ob). Using a preliminary cohort of normal human subjects, we found the thickness of skin-associated fat varied 8-fold, from 0.13-1.10 cm; on average, this depot is calculated to weigh 8.8 kg.
Collapse
Affiliation(s)
| | | | | | | | - Scott B Reeder
- Department of Radiology.,Department of Medical Physics.,Department of Biomedical Engineering.,Department of Medicine, and.,Department of Emergency Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
21
|
Ibrahim SA, Hassan H, Götte M. MicroRNA regulation of proteoglycan function in cancer. FEBS J 2014; 281:5009-22. [DOI: 10.1111/febs.13026] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/08/2014] [Accepted: 08/26/2014] [Indexed: 01/08/2023]
Affiliation(s)
- Sherif A. Ibrahim
- Department of Zoology; Faculty of Science; Cairo University; Giza Egypt
| | - Hebatallah Hassan
- Department of Zoology; Faculty of Science; Cairo University; Giza Egypt
| | - Martin Götte
- Department of Gynecology and Obstetrics; Münster University Hospital; Germany
| |
Collapse
|
22
|
Kasza I, Suh Y, Wollny D, Clark RJ, Roopra A, Colman RJ, MacDougald OA, Shedd TA, Nelson DW, Yen MI, Yen CLE, Alexander CM. Syndecan-1 is required to maintain intradermal fat and prevent cold stress. PLoS Genet 2014; 10:e1004514. [PMID: 25101993 PMCID: PMC4125098 DOI: 10.1371/journal.pgen.1004514] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 06/05/2014] [Indexed: 02/07/2023] Open
Abstract
Homeostatic temperature regulation is fundamental to mammalian physiology and is controlled by acute and chronic responses of local, endocrine and nervous regulators. Here, we report that loss of the heparan sulfate proteoglycan, syndecan-1, causes a profoundly depleted intradermal fat layer, which provides crucial thermogenic insulation for mammals. Mice without syndecan-1 enter torpor upon fasting and show multiple indicators of cold stress, including activation of the stress checkpoint p38α in brown adipose tissue, liver and lung. The metabolic phenotype in mutant mice, including reduced liver glycogen, is rescued by housing at thermoneutrality, suggesting that reduced insulation in cool temperatures underlies the observed phenotypes. We find that syndecan-1, which functions as a facultative lipoprotein uptake receptor, is required for adipocyte differentiation in vitro. Intradermal fat shows highly dynamic differentiation, continuously expanding and involuting in response to hair cycle and ambient temperature. This physiology probably confers a unique role for Sdc1 in this adipocyte sub-type. The PPARγ agonist rosiglitazone rescues Sdc1−/− intradermal adipose tissue, placing PPARγ downstream of Sdc1 in triggering adipocyte differentiation. Our study indicates that disruption of intradermal adipose tissue development results in cold stress and complex metabolic pathology. All mammals strive to maintain a fixed body temperature, and do so using a remarkable array of different strategies, which vary depending upon the degree of cold challenge. Physiologists many decades ago observed that a fat layer right underneath the epidermis (and above the dermal muscle layer) thickens in response to colder ambient temperatures. This “intradermal fat” provided insulation within days of climate changes. We have found that syndecan-1, which functions as a facultative lipoprotein uptake receptor, is required for intradermal fat expansion in response to cold exposure. This is a highly specific phenotype not shared by other adipocytes. When intradermal fat is absent, mice do not adapt normally to cold stress, and show altered systemic physiologies, including increased brown adipose tissue thermogenesis and hyper-activation of a stress checkpoint (p38α), designed to protect the body against mutagenic and oxidative stressors. The phenotypes associated with loss of Sdc1 function are reversed when mice are housed in warm temperatures, where defense of body temperature is not required. This study is the first to show that intradermal fat can be genetically regulated, with systemic effects on physiology.
Collapse
Affiliation(s)
- Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yewseok Suh
- Department of Dermatology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Damian Wollny
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rod J. Clark
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ricki J. Colman
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Timothy A. Shedd
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David W. Nelson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mei-I Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chi-Liang Eric Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
23
|
Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, Schneider C, Kiesel L, Eich HT, Zucchi I, Reinbold R, Greve B, Götte M. Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-mediated STAT3 signaling. PLoS One 2013; 8:e85737. [PMID: 24392029 PMCID: PMC3877388 DOI: 10.1371/journal.pone.0085737] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/02/2013] [Indexed: 12/17/2022] Open
Abstract
Syndecan-1 (CD138), a heparan sulfate proteoglycan, acts as a coreceptor for growth factors and chemokines and is a molecular marker associated with epithelial-mesenchymal transition during development and carcinogenesis. Resistance of Syndecan-1-deficient mice to experimentally-induced tumorigenesis has been linked to altered Wnt-responsive precursor cell pools, suggesting a potential role of Syndecan-1 in breast cancer cell stem function. However, the precise molecular mechanism is still elusive. Here, we decipher the functional impact of Syndecan-1 knockdown using RNA interference on the breast cancer stem cell phenotype of human triple-negative MDA-MB-231 and hormone receptor-positive MCF-7 cells in vitro employing an analytical flow cytometric approach. Successful Syndecan-1 siRNA knockdown was confirmed by flow cytometry. Side population measurement by Hoechst dye exclusion and Aldehyde dehydrogenase-1 activity revealed that Syndecan-1 knockdown in MDA-MB-231 cells significantly reduced putative cancer stem cell pools by 60% and 27%, respectively, compared to controls. In MCF-7 cells, Syndecan-1 depletion reduced the side population by 40% and Aldehyde dehydrogenase-1 by 50%, repectively. In MDA-MB-231 cells, the CD44(+)CD24(-/low) phenotype decreased significantly by 6% upon siRNA-mediated Syndecan-1 depletion. Intriguingly, IL-6, its receptor sIL-6R, and the chemokine CCL20, implicated in regulating stemness-associated pathways, were downregulated by >40% in Syndecan-1-silenced MDA-MB-231 cells, which showed a dysregulated response to IL-6-induced shifts in E-cadherin and vimentin expression. Furthermore, activation of STAT-3 and NFkB transcription factors and expression of a coreceptor for Wnt signaling, LRP-6, were reduced by >45% in Syndecan-1-depleted cells compared to controls. At the functional level, Syndecan-1 siRNA reduced the formation of spheres and cysts in MCF-7 cells grown in suspension culture. Our study demonstrates the viability of flow cytometric approaches in analyzing cancer stem cell function. As Syndecan-1 modulates the cancer stem cell phenotype via regulation of the Wnt and IL-6/STAT3 signaling pathways, it emerges as a promising novel target for therapeutic approaches.
Collapse
Affiliation(s)
- Sherif A Ibrahim
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany ; Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Hebatallah Hassan
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany ; Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Sampath Katakam Kumar
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Archana Vijaya Kumar
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Reinhard Kelsch
- Institute of Transfusion Medicine and Transplantation Immunology, University Hospital Münster, Münster, Germany
| | - Cornelia Schneider
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Hans Theodor Eich
- Department of Radiotherapy - Radiooncology, University Hospital Münster, Münster, Germany
| | | | | | - Burkhard Greve
- Department of Radiotherapy - Radiooncology, University Hospital Münster, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| |
Collapse
|
24
|
Zhang X, Wu C, Song J, Götte M, Sorokin L. Syndecan-1, a cell surface proteoglycan, negatively regulates initial leukocyte recruitment to the brain across the choroid plexus in murine experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 191:4551-61. [PMID: 24078687 DOI: 10.4049/jimmunol.1300931] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The cell surface heparan sulfate proteoglycan, syndecan-1, has been reported to be a negative regulator of various inflammatory processes, but its precise mode of action is poorly defined. In this study, we use the murine model of the 35-55 peptide of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE), a T lymphocyte-mediated inflammation where the steps in disease development and recovery are well characterized, to decipher how syndecan-1 impacts on the inflammatory reaction. Syndecan-1 knockout (Sdc-1(-/-)) mice show enhanced disease severity and impaired recovery. The use of bone marrow chimeric mice reveals that both an immune cell and a CNS-resident source of syndecan-1 contribute to this phenotype. Epithelial cells of the choroid plexus, where initial CCL20-induced leukocyte recruitment to the brain occurs, are identified as the predominant site of syndecan-1 expression. Syndecan-1 is lost from this site during the course of EAE by shedding into the cerebrospinal fluid, which correlates with loss of epithelial cell surface-bound CCL20 and is associated with the upregulation of IL-6 expression. In Sdc-1(-/-) mice, early leukocyte recruitment via the choroid plexus is enhanced, and IL-6 is elevated, which collectively results in higher numbers of the disease inducing Th17 cells in the CNS, thereby contributing to enhanced disease severity. Furthermore, Sdc-1(-/-) mice have intrinsically elevated plasma cell numbers and higher myelin oligodendrocyte glycoprotein-specific Ab levels during EAE, which we propose contributes to impaired recovery. Our data identify the choroid plexus epithelium as a novel source of IL-6 in EAE and demonstrate that its expression negatively correlates with syndecan-1 expression at this site.
Collapse
Affiliation(s)
- Xueli Zhang
- Institute for Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | | | | | | | | |
Collapse
|
25
|
Liu C, Gu S, Sun C, Ye W, Song Z, Zhang Y, Chen Y. FGF signaling sustains the odontogenic fate of dental mesenchyme by suppressing β-catenin signaling. Development 2013; 140:4375-85. [PMID: 24067353 DOI: 10.1242/dev.097733] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Odontoblasts and osteoblasts develop from multipotent craniofacial neural crest cells during tooth and jawbone development, but the mechanisms that specify and sustain their respective fates remain largely unknown. In this study we used early mouse molar and incisor tooth germs that possess distinct tooth-forming capability after dissociation and reaggregation in vitro to investigate the mechanism that sustains odontogenic fate of dental mesenchyme during tooth development. We found that after dissociation and reaggregation, incisor, but not molar, mesenchyme exhibits a strong osteogenic potency associated with robustly elevated β-catenin signaling activity in a cell-autonomous manner, leading to failed tooth formation in the reaggregates. Application of FGF3 to incisor reaggregates inhibits β-catenin signaling activity and rescues tooth formation. The lack of FGF retention on the cell surface of incisor mesenchyme appears to account for the differential osteogenic potency between incisor and molar, which can be further attributed to the differential expression of syndecan 1 and NDST genes. We further demonstrate that FGF signaling inhibits intracellular β-catenin signaling by activating the PI3K/Akt pathway to regulate the subcellular localization of active GSK3β in dental mesenchymal cells. Our results reveal a novel function for FGF signaling in ensuring the proper fate of dental mesenchyme by regulating β-catenin signaling activity during tooth development.
Collapse
Affiliation(s)
- Chao Liu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Schmedt A, Götte M, Heinig J, Kiesel L, Klockenbusch W, Steinhard J. Evaluation of placental syndecan-1 expression in early pregnancy as a predictive fetal factor for pregnancy outcome. Prenat Diagn 2012; 32:131-7. [PMID: 22418956 DOI: 10.1002/pd.2908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Early expression of syndecan-1, a regulator of growth factor and chemokine function, in animal and human abortuses or placenta at delivery suggests a potential role during embryonic development. The aim of this study was to examine the correlation between early placental syndecan expression and fetal outcome in ongoing pregnancies. METHODS Prospective immunohistochemical analysis of 203 chorionic villus sampling specimens obtained during a 4-year period (2001-2004). The chorionic villi were processed for paraffin-embedded immunohistochemistry. Scores of intensity of syndecan-1 expression in chorionic villi were correlated with maternal cofactors and pregnancy outcome. RESULTS Syndecan-1 immunostaining was significantly related to villus size, maternal tobacco use, and fetal biometry at ultrasonography. After correcting the above confounders, scores of syndecan-1 expression correlated with preterm birth and low birth weight. CONCLUSIONS Placental syndecan-1 expression is associated with premature birth and fetal growth.
Collapse
Affiliation(s)
- Anna Schmedt
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Str. 33, D-48149, Münster, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Huang Y, Jiang W, Wang Y, Zheng Y, Cong Q, Xu C. Enhanced efficacy and specificity of epithelial ovarian carcinogenesis by embedding a DMBA-coated cloth strip in the ovary of rat. J Ovarian Res 2012; 5:21. [PMID: 22943261 PMCID: PMC3479048 DOI: 10.1186/1757-2215-5-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/21/2012] [Indexed: 11/27/2022] Open
Abstract
Background Ovarian cancer is predominant of epithelial cell origin and often present at an advanced stage with poor prognosis. Most animal models of ovarian carcinoma yield thecal/granulose cell tumors, rather than adenocarcinomas. The best reported induction rate of adenocarcinoma in rats is 10-45% by an ovarian implantation of 7, 12-dimethylbenz[a]anthracene (DMBA) coated silk suture. We provided an improved procedure to construct the model by the ovarian implantation of DMBA-coated cloth strip. Methods A sterile suture (as S group) or a piece of cloth strip (as CS group) was soaked in DMBA before ovarian implantation in Wistar rats. Tumor size, incidence rate and pathological type were analyzed. Results Ovarian tumors in rats of CS group were first noted at 16 wk post implantation and reached a cumulative incidence of 75% (96/128) at 32 wk, while the tumor incidence rate in S group at 32 wk was only 46.25% (37/80). The tumor size in CS group (3.63 ± 0.89 cm) was larger than that of S group (2.44 ± 1.89 cm) (P < 0.05). In CS group, there were only two types of tumor formed: adenocarcinoma (90/96) and sarcoma (6/96). While in S group, there were different types, including adenocarcinoma (21/37), squamous carcinoma (3/37), granulosa cell tumor (3/37), sarcoma (4/37), undifferentiated carcinoma with no adeno character (2/37), benign ovarian tumor (2/37), and malignant teratoma (1/37). Conclusion The model in our study yields much higher incidence and specificity of epithelial derived tumors and showed histological similarities to human ovarian cancers, which would be more suitable for therapeutic research.
Collapse
Affiliation(s)
- Yiping Huang
- Obstetrics and Gynecology Hospital, Department of Obstetrics and Gynecology of Shanghai Medical School, and Institute of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
28
|
Wang Q, Yang L, Alexander C, Temple S. The niche factor syndecan-1 regulates the maintenance and proliferation of neural progenitor cells during mammalian cortical development. PLoS One 2012; 7:e42883. [PMID: 22936997 PMCID: PMC3427302 DOI: 10.1371/journal.pone.0042883] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/13/2012] [Indexed: 12/18/2022] Open
Abstract
Neural progenitor cells (NPCs) divide and differentiate in a precisely regulated manner over time to achieve the remarkable expansion and assembly of the layered mammalian cerebral cortex. Both intrinsic signaling pathways and environmental factors control the behavior of NPCs during cortical development. Heparan sulphate proteoglycans (HSPG) are critical environmental regulators that help modulate and integrate environmental cues and downstream intracellular signals. Syndecan-1 (Sdc1), a major transmembrane HSPG, is highly enriched in the early neural germinal zone, but its function in modulating NPC behavior and cortical development has not been explored. In this study we investigate the expression pattern and function of Sdc1 in the developing mouse cerebral cortex. We found that Sdc1 is highly expressed by cortical NPCs. Knockdown of Sdc1 in vivo by in utero electroporation reduces NPC proliferation and causes their premature differentiation, corroborated in isolated cells in vitro. We found that Sdc1 knockdown leads to reduced levels of β-catenin, indicating reduced canonical Wnt signaling. Consistent with this, GSK3β inhibition helps rescue the Sdc1 knockdown phenotype, partially restoring NPC number and proliferation. Moreover, exogenous Wnt protein promotes cortical NPC proliferation, but this is prevented by Sdc1 knockdown. Thus, Sdc1 in the germinal niche is a key HSPG regulating the maintenance and proliferation of NPCs during cortical neurogenesis, in part by modulating the ability of NPCs to respond to Wnt ligands.
Collapse
Affiliation(s)
- Qingjie Wang
- Neural Stem Cell Institute, Rensselaer, New York, United States of America
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York, United States of America
| | - Landi Yang
- Neural Stem Cell Institute, Rensselaer, New York, United States of America
| | - Caroline Alexander
- McArdle Lab for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, New York, United States of America
- * E-mail:
| |
Collapse
|
29
|
Ibrahim SA, Yip GW, Stock C, Pan JW, Neubauer C, Poeter M, Pupjalis D, Koo CY, Kelsch R, Schüle R, Rescher U, Kiesel L, Götte M. Targeting of syndecan-1 by microRNA miR-10b promotes breast cancer cell motility and invasiveness via a Rho-GTPase- and E-cadherin-dependent mechanism. Int J Cancer 2012; 131:E884-96. [PMID: 22573479 DOI: 10.1002/ijc.27629] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/25/2012] [Indexed: 12/22/2022]
Abstract
microRNAs are small endogenous noncoding RNAs, which post-transcriptionally regulate gene expression. In breast cancer, overexpression of the transmembrane heparan sulfate proteoglycan syndecan-1, a predicted target of the oncomiR miR-10b, correlates with poor clinical outcome. To investigate the potential functional relationship of miR-10b and syndecan-1, MDA-MB-231 and MCF-7 breast cancer cells were transiently transfected with pre-miR-10b, syndecan-1 siRNA or control reagents, respectively. Altered cell behavior was monitored by proliferation, migration and invasion chamber assays, and time-lapse video microscopy. miR-10b overexpression induced post-transcriptional downregulation of syndecan-1, as demonstrated by quantitative real-time PCR (qPCR), flow cytometry, and 3'UTR luciferase assays, resulting in increased cancer cell migration and matrigel invasiveness. Syndecan-1 silencing generated a copy of this phenotype. Adhesion to fibronectin and laminin and basal cell proliferation was increased. Syndecan-1 coimmunoprecipitated with focal adhesion kinase, which showed increased activation upon syndecan-1 depletion. Affymetrix screening and confirmatory qPCR and Western blotting analysis of syndecan-1-deficient cells revealed upregulation of ATF-2, COX-2, cadherin-11, vinculin, actin γ 2, MYL9, transgelin-1, RhoA/C, matrix metalloproteinase 2 (MMP2) and heparanase, and downregulation of AML1/RUNX1, E-cadherin, CLDN1, p21WAF/CIP, cyclin-dependent kinase 6, TLR-4, PAI1/2, Collagen1alpha1, JHDM1D, Mpp4, MMP9, matrilin-2 and ANXA3/A10. Video microscopy demonstrated massively increased Rho kinase-dependent motility of syndecan-1-depleted cells, which displayed increased filopodia formation. We conclude that syndecan-1 is a novel target of the oncomiR miR-10b. Rho-GTPase-dependent modulation of cytoskeletal function and downregulation of E-cadherin expression are identified as relevant effectors of the miR-10b-syndecan-1 axis, which emerges as a promising target for the development of new therapeutic approaches for breast cancer.
Collapse
Affiliation(s)
- Sherif A Ibrahim
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim S, Roopra A, Alexander CM. A phenotypic mouse model of basaloid breast tumors. PLoS One 2012; 7:e30979. [PMID: 22347416 PMCID: PMC3276569 DOI: 10.1371/journal.pone.0030979] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 12/30/2011] [Indexed: 12/23/2022] Open
Abstract
Chemotherapeutic strategies that target basal-like breast tumors are difficult to design without understanding their cellular and molecular basis. Here, we induce tumors in mice by carcinogen administration, creating a phenocopy of tumors with the diagnostic and functional aspects of human triple negative disease (including EGFR expression/lack of erbB, estrogen-independent growth and co-clustering of the transcriptome with other basaloid models). These tumor strains are a complement to established mouse models that are based on mutations in Brca1 and/or p53. Tumors comprise two distinct cell subpopulations, basal and luminal epithelial cells. These cell fractions were purified by flow cytometry, and only basal cell fractions found to have tumor initiating activity (cancer stem cells). The phenotype of serially regenerated tumors was stable, and irrespective of tumor precursor cell. Tumors were passaged entirely in vivo and serial generations tested for their phenotypic stability. The relative chemo-sensitivity of basal and luminal cells were evaluated. Upon treatment with anthracycline, tumors were effectively de-bulked, but recurred; this correlated with maintenance of a high rate of basal cell division throughout the treatment period. Thus, these tumors grow as robust cell mixtures of basal bipotential tumor initiating cells alongside a luminal majority, and the cellular response to drug administration is dominated by the distinct biology of the two cell types. Given the ability to separate basal and luminal cells, and the discovery potential of this approach, we propose that this mouse model could be a convenient one for preclinical studies.
Collapse
Affiliation(s)
- Soyoung Kim
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
31
|
Garusi E, Rossi S, Perris R. Antithetic roles of proteoglycans in cancer. Cell Mol Life Sci 2012; 69:553-79. [PMID: 21964924 PMCID: PMC11114698 DOI: 10.1007/s00018-011-0816-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 09/01/2011] [Accepted: 09/05/2011] [Indexed: 12/15/2022]
Abstract
Proteoglycans (PGs), a family of complex post-translationally sculptured macromolecules, are fundamental regulators of most normal and aberrant cellular functions. The unparalleled structural-functional diversity of PGs endows them with the ability to serve as critical mediators of the tumor cells' interaction with the host microenvironment, while directly contributing to the organization and dynamic remodeling of this milieu. Despite their indisputable importance during embryonic development and in the adult organism, and their frequent dysregulation in tumor lesions, their precise involvement in tumorigenesis awaits a more decisive demonstration. Particularly challenging is to ascertain to what extent selected PGs may catalyze tumor progression and to what extent they may inhibit it, implying antithetic functions of individual PGs. Integrated efforts are needed to consolidate the routine use of PGs in the clinical monitoring of cancer patients and to broaden the exploitation of these macromolecules as therapeutic targets. Several PGs have the required attributes to be contemplated as effective antigens for immunotherapeutic approaches, while the tangible results obtained in recent clinical trials targeting the NG2/CSPG4 transmembrane PG urge further development of PG-based cancer treatment modalities.
Collapse
Affiliation(s)
- Elena Garusi
- COMT, Centre for Molecular and Translational Oncology, University of Parma, Via G.P. Usberti 11/A, 43100 Parma, Italy
| | - Silvia Rossi
- COMT, Centre for Molecular and Translational Oncology, University of Parma, Via G.P. Usberti 11/A, 43100 Parma, Italy
- Department of Genetic, Biology of Microorganism, Anthropology and Evolution, University of Parma, Via G.P. Usberti 11/A, 43100 Parma, Italy
| | - Roberto Perris
- COMT, Centre for Molecular and Translational Oncology, University of Parma, Via G.P. Usberti 11/A, 43100 Parma, Italy
- Department of Genetic, Biology of Microorganism, Anthropology and Evolution, University of Parma, Via G.P. Usberti 11/A, 43100 Parma, Italy
- S.O.C. of Experimental Oncology 2, The National Cancer Institute Aviano, CRO-IRCCS, Via Franco Gallini, 2, 33081 Aviano, PN Italy
| |
Collapse
|
32
|
Seidler DG, Mohamed NA, Bocian C, Stadtmann A, Hermann S, Schäfers K, Schäfers M, Iozzo RV, Zarbock A, Götte M. The role for decorin in delayed-type hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2011; 187:6108-19. [PMID: 22043007 DOI: 10.4049/jimmunol.1100373] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Decorin, a small leucine-rich proteoglycan, regulates extracellular matrix organization, growth factor-mediated signaling, and cell growth. Because decorin may directly modulate immune responses, we investigated its role in a mouse model of contact allergy (oxazolone-mediated delayed-type hypersensitivity [DTH]) in decorin-deficient (Dcn(-/-)) and wild-type mice. Dcn(-/-) mice showed a reduced ear swelling 24 h after oxazolone treatment with a concurrent attenuation of leukocyte infiltration. These findings were corroborated by reduced glucose metabolism, as determined by (18)fluordeoxyglucose uptake in positron emission tomography scans. Unexpectedly, polymorphonuclear leukocyte numbers in Dcn(-/-) blood vessels were significantly increased and accompanied by large numbers of flattened leukocytes adherent to the endothelium. Intravital microscopy and flow chamber and static adhesion assays confirmed increased adhesion and reduced transmigration of Dcn(-/-) leukocytes. Circulating blood neutrophil numbers were significantly increased in Dcn(-/-) mice 24 h after DTH elicitation, but they were only moderately increased in wild-type mice. Expression of the proinflammatory cytokine TNF-α was reduced, whereas syndecan-1 and ICAM-1 were overexpressed in inflamed ears of Dcn(-/-) mice, indicating that these adhesion molecules could be responsible for increased leukocyte adhesion. Decorin treatment of endothelial cells increased tyrosine phosphorylation and reduced syndecan-1 expression. Notably, absence of syndecan-1 in a genetic background lacking decorin rescued the attenuated DTH phenotype of Dcn(-/-) mice. Collectively, these results implicated a role for decorin in mediating DTH responses by influencing polymorphonuclear leukocyte attachment to the endothelium. This occurs via two nonmutually exclusive mechanisms that involve a direct antiadhesive effect on polymorphonuclear leukocytes and a negative regulation of ICAM-1 and syndecan-1 expression.
Collapse
Affiliation(s)
- Daniela G Seidler
- Institute of Physiological Chemistry and Pathobiochemistry, D-48149 Münster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Keely PJ. Mechanisms by which the extracellular matrix and integrin signaling act to regulate the switch between tumor suppression and tumor promotion. J Mammary Gland Biol Neoplasia 2011; 16:205-19. [PMID: 21822945 PMCID: PMC3885166 DOI: 10.1007/s10911-011-9226-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 07/19/2011] [Indexed: 12/21/2022] Open
Abstract
Cell adhesion to the extracellular matrix (ECM) is necessary for development of the mammary gland, and to maintain the normal architecture and function of the gland. Cells adhere to the ECM via the integrin family of trans-membrane receptors, which signal to control mammary-specific gene expression and regulate cell proliferation and survival. During tumor formation, the ECM is extensively remodeled and signaling through integrins is altered such that cells become proliferative and invasive. A key regulator of whether integrin-mediated adhesion will promote tumor suppression or tumor formation is the stiffness of the stromal ECM. The normal mammary gland is typically surrounded by a loose collagenous stroma. An increase in the deposition of collagen and other stromal components is associated with mammographic density, which is one of the greatest risk factors for developing breast carcinoma. Several groups have demonstrated that increased stromal ECM density results in a matrix that is stiffer. Cells sense the stiffness of their surrounding ECM by Rho-mediated contraction of the actin-myosin cytoskeleton. If the surrounding ECM is stiffer than the cell's ability to contract it, then the tensile forces that result are able to drive the clustering of integrins and assemble adhesion signaling complexes. The result is subsequent activation of signaling pathways including FAK, ERK, and PI3K that drive cell proliferation and survival. In contrast, focal complexes are not formed in a compliant matrix, and activation of FAK and pERK is diminished, resulting in control of proliferation. Signaling from FAK moreover regulates p53 and miR-200 members, which control apoptosis and epithelial phenotype, such that a compliant matrix is predicted to promote normal mammary gland architecture and suppress tumor formation.
Collapse
Affiliation(s)
- Patricia J Keely
- Department of Cell and Regenerative Biology, Laboratory for Cellular and Molecular Biology, & Laboratory for Optical and Computational Instrumentation, University of Wisconsin, 227D Bock Laboratories, 1525 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
34
|
Sarrazin S, Lamanna WC, Esko JD. Heparan sulfate proteoglycans. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a004952. [PMID: 21690215 DOI: 10.1101/cshperspect.a004952] [Citation(s) in RCA: 1091] [Impact Index Per Article: 77.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heparan sulfate proteoglycans are found at the cell surface and in the extracellular matrix, where they interact with a plethora of ligands. Over the last decade, new insights have emerged regarding the mechanism and biological significance of these interactions. Here, we discuss changing views on the specificity of protein-heparan sulfate binding and the activity of HSPGs as receptors and coreceptors. Although few in number, heparan sulfate proteoglycans have profound effects at the cellular, tissue, and organismal level.
Collapse
Affiliation(s)
- Stephane Sarrazin
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
35
|
Abstract
Heparan sulfate proteoglycans are found at the cell surface and in the extracellular matrix, where they interact with a plethora of ligands. Over the last decade, new insights have emerged regarding the mechanism and biological significance of these interactions. Here, we discuss changing views on the specificity of protein-heparan sulfate binding and the activity of HSPGs as receptors and coreceptors. Although few in number, heparan sulfate proteoglycans have profound effects at the cellular, tissue, and organismal level.
Collapse
Affiliation(s)
- Stephane Sarrazin
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
36
|
Sulkowski GN, Warren J, Ha CT, Dveksler GS. Characterization of receptors for murine pregnancy specific glycoproteins 17 and 23. Placenta 2011; 32:603-10. [PMID: 21669460 DOI: 10.1016/j.placenta.2011.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 05/16/2011] [Accepted: 05/18/2011] [Indexed: 10/18/2022]
Abstract
In primates and rodents, trophoblast cells synthesize and secrete into the maternal circulation a family of proteins known as pregnancy specific glycoproteins (PSG). The current study was undertaken to characterize the receptor for two members of the murine PSG family, PSG17 and PSG23. Binding of recombinant PSG17 and PSG23 to CHO-K1 and L929 cells and their derived mutants was performed to determine whether these proteins bound to cell surface proteoglycans. We also examined binding of these proteins to cells transfected with syndecans and glypican-1 by flow cytometry. The interaction with glycosaminoglycans was confirmed in solid phase assays. Our results show that PSG17 binds to CD9 and to cell surface proteoglycans while PSG23 binds only to the latter. We found that the amino acids involved in CD9 binding reside in the region of highest divergence between the N1-domains of murine PSGs. For both proteins, the N-terminal domain (designated as N1) is sufficient for binding to cells and the ability to bind cell surface proteoglycans is affected by the cell line employed to generate the recombinant proteins. We conclude that while substantially different at the amino acid level, some murine PSGs share with human PSG1 the ability to bind to cell surface proteoglycans and that at least one PSG binds to more than one type of molecule on the cell surface.
Collapse
Affiliation(s)
- G N Sulkowski
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | |
Collapse
|
37
|
Conklin MW, Eickhoff JC, Riching KM, Pehlke CA, Eliceiri KW, Provenzano PP, Friedl A, Keely PJ. Aligned collagen is a prognostic signature for survival in human breast carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1221-32. [PMID: 21356373 DOI: 10.1016/j.ajpath.2010.11.076] [Citation(s) in RCA: 949] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 11/11/2010] [Accepted: 11/17/2010] [Indexed: 12/11/2022]
Abstract
Evidence for the potent influence of stromal organization and function on invasion and metastasis of breast tumors is ever growing. We have performed a rigorous examination of the relationship of a tumor-associated collagen signature-3 (TACS-3) to the long-term survival rate of human patients. TACS-3 is characterized by bundles of straightened and aligned collagen fibers that are oriented perpendicular to the tumor boundary. An evaluation of TACS-3 was performed in biopsied tissue sections from 196 patients by second harmonic generation imaging of the backscattered signal generated by collagen. Univariate analysis of a Cox proportional hazard model demonstrated that the presence of TACS-3 was associated with poor disease-specific and disease-free survival, resulting in hazard ratios between 3.0 and 3.9. Furthermore, TACS-3 was confirmed to be an independent prognostic indicator regardless of tumor grade and size, estrogen or progesterone receptor status, human epidermal growth factor receptor-2 status, node status, and tumor subtype. Interestingly, TACS-3 was positively correlated to expression of stromal syndecan-1, a receptor for several extracellular matrix proteins including collagens. Because of the strong statistical evidence for poor survival in patients with TACS, and because the assessment can be performed in routine histopathological samples imaged via second harmonic generation or using picrosirius, we propose that quantifying collagen alignment is a viable, novel paradigm for the prediction of human breast cancer survival.
Collapse
Affiliation(s)
- Matthew W Conklin
- Department of Pharmacology, and the Laboratory for Molecular Biology, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Iozzo RV, Sanderson RD. Proteoglycans in cancer biology, tumour microenvironment and angiogenesis. J Cell Mol Med 2011; 15:1013-31. [PMID: 21155971 PMCID: PMC3633488 DOI: 10.1111/j.1582-4934.2010.01236.x] [Citation(s) in RCA: 434] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 12/08/2010] [Indexed: 12/12/2022] Open
Abstract
Proteoglycans, key molecular effectors of cell surface and pericellular microenvironments, perform multiple functions in cancer and angiogenesis by virtue of their polyhedric nature and their ability to interact with both ligands and receptors that regulate neoplastic growth and neovascularization. Some proteoglycans such as perlecan, have pro- and anti-angiogenic activities, whereas other proteoglycans, such as syndecans and glypicans, can also directly affect cancer growth by modulating key signalling pathways. The bioactivity of these proteoglycans is further modulated by several classes of enzymes within the tumour microenvironment: (i) sheddases that cleave transmembrane or cell-associated syndecans and glypicans, (ii) various proteinases that cleave the protein core of pericellular proteoglycans and (iii) heparanases and endosulfatases which modify the structure and bioactivity of various heparan sulphate proteoglycans and their bound growth factors. In contrast, some of the small leucine-rich proteoglycans, such as decorin and lumican, act as tumour repressors by physically antagonizing receptor tyrosine kinases including the epidermal growth factor and the Met receptors or integrin receptors thereby evoking anti-survival and pro-apoptotic pathways. In this review we will critically assess the expanding repertoire of molecular interactions attributed to various proteoglycans and will discuss novel proteoglycan functions modulating cancer progression, invasion and metastasis and how these factors regulate the tumour microenvironment.
Collapse
Affiliation(s)
- Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson UniversityPhiladelphia, PA, USA
| | - Ralph D Sanderson
- Department of Pathology, and the Comprehensive Cancer Center, University of Alabama at BirminghamBirmingham, AL, USA
| |
Collapse
|
39
|
Baston-Büst DM, Götte M, Janni W, Krüssel JS, Hess AP. Syndecan-1 knock-down in decidualized human endometrial stromal cells leads to significant changes in cytokine and angiogenic factor expression patterns. Reprod Biol Endocrinol 2010; 8:133. [PMID: 21044331 PMCID: PMC2988802 DOI: 10.1186/1477-7827-8-133] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 11/02/2010] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Successful embryonic implantation depends on a synchronized embryo-maternal dialogue. Chemokines, such as chemokine ligand 1 (CXCL1), play essential roles in the maternal reproductive tract leading to morphological changes during decidualization, mediating maternal acceptance towards the semi-allograft embryo and induction of angiogenesis. Chemokine binding to their classical G-protein coupled receptors is essentially supported by the syndecan (Sdc) family of heparan sulfate proteoglycans. The aim of this study was to identify the involvement of Sdc-1 at the embryo-maternal interface regarding changes of the chemokine and angiogenic profile of the decidua during the process of decidualization and implantation in human endometrium. METHODS A stable Sdc-1 knock-down was generated in the immortalized human endometrial stromal cell line St-T1 and was named KdS1. The ability of KdS1 to decidualize was proven by Insulin-like growth factor binding 1 (IGFBP1) and prolactin (PRL) confirmation on mRNA level before further experiments were carried out. Dot blot protein analyses of decidualized knock-down cells vs non-transfected controls were performed. In order to imitate embryonic implantation, decidualized KdS1 were then incubated with IL-1beta, an embryo secretion product, vs controls. Statistical analyses were performed applying the Student's t-test with p < 0.05, p < 0.02 and p < 0.01 and one way post-hoc ANOVA test with p < 0.05 as cut-offs for statistical significance. RESULTS The induction of the Sdc-1 knock-down revealed significant changes in cytokine and angiogenic factor expression profiles of dKdS1 vs decidualized controls. Incubation with embryonic IL-1beta altered the expression patterns of KdS1 chemokines and angiogenic factors towards inflammatory-associated molecules and factors involved in matrix regulation. CONCLUSIONS Sdc-1 knock-down in human endometrial stroma cells led to fulminant changes regarding cytokine and angiogenic factor expression profiles upon decidualization and imitation of embryonic contact. Sdc-1 appears to play an important role as a co-receptor and storage factor for many cytokines and angiogenic factors during decidualization and implantation period, supporting proper implantation and angiogenesis by regulation of chemokine and angiogenic factor secretion in favour of the implanting embryo.
Collapse
Affiliation(s)
- Dunja M Baston-Büst
- University Düsseldorf, Medical Faculty, Department of OB/GYN and REI, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Martin Götte
- Department of OB/GYN, Münster University Hospital, Albert-Schweitzer-Str. 33, 48149 Münster, Germany
| | - Wolfgang Janni
- University Düsseldorf, Medical Faculty, Department of OB/GYN and REI, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Jan-Steffen Krüssel
- University Düsseldorf, Medical Faculty, Department of OB/GYN and REI, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Alexandra P Hess
- University Düsseldorf, Medical Faculty, Department of OB/GYN and REI, Moorenstr. 5, 40225 Düsseldorf, Germany
| |
Collapse
|
40
|
Stepp MA, Pal-Ghosh S, Tadvalkar G, Rajjoub L, Jurjus RA, Gerdes M, Ryscavage A, Cataisson C, Shukla A, Yuspa SH. Loss of syndecan-1 is associated with malignant conversion in skin carcinogenesis. Mol Carcinog 2010; 49:363-73. [PMID: 20082322 DOI: 10.1002/mc.20609] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Syndecan-1 (sdc-1) is a cell surface proteoglycan that mediates the interaction of cells with their matrix, influencing attachment, migration, and response to growth factors. In keratinocytes, loss of sdc-1 delays wound healing, reduces migration, and increases Transforming growth factor beta (TGFbeta) 1 expression. In this study we show that sdc-1 expression is significantly reduced in basal cell, squamous cell, and metastatic human skin cancers compared to normal human skin. In experimental mouse skin tumor induction, compared to wildtype (wt) BALB/c mice, papilloma formation in sdc-1 null mice was reduced by 50% and the percent of papillomas converting to squamous cell carcinoma (SCC) was enhanced. sdc-1 expression on wt mouse papillomas decreased as they converted to SCC. Furthermore, papillomas forming on sdc-1 null mice expressed suprabasal alpha3 and beta4 integrins; suprabasal beta4 integrin is a marker of a high risk for progression. While the proliferative response to phorbol-12-myristate-13-acetate (TPA) did not differ among the genotypes, sdc-1 null mice had an enhanced inflammatory response and retained higher levels of total TGFbeta1 within their skin after TPA treatment. sdc-1 null keratinocytes, transduced in vitro by oncogenic ras(Ha), expressed higher levels of beta4 integrin and had enhanced pSmad2 signaling and reduced senescence when compared to wt ras(Ha)-transduced keratinocytes. When ras(Ha)-transduced cells of both genotypes were grafted onto nude mice, null tumors converted to SCC with higher frequency confirming the skin painting experiments. These data indicate that sdc-1 is important both early in the development of skin tumors and in progression of skin cancers suggesting that reduced expression of sdc-1 could be a useful marker for progression in neoplastic skin lesions.
Collapse
Affiliation(s)
- Mary Ann Stepp
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, District of Columbia, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Götte M. Endometrial cells get side-tracked: side population cells promote epithelial-mesenchymal transition in endometrial carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:25-8. [PMID: 19948831 DOI: 10.2353/ajpath.2010.090775] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Str. 33, D-48149 Münster, Germany.
| |
Collapse
|
42
|
Kirn-Safran C, Farach-Carson MC, Carson DD. Multifunctionality of extracellular and cell surface heparan sulfate proteoglycans. Cell Mol Life Sci 2009; 66:3421-34. [PMID: 19629389 PMCID: PMC11115568 DOI: 10.1007/s00018-009-0096-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 07/01/2009] [Accepted: 07/06/2009] [Indexed: 02/06/2023]
Abstract
Heparan sulfate proteoglycans are a remarkably diverse family of glycosaminoglycan-bearing protein cores that include the syndecans, the glypicans, perlecan, agrin, and collagen XVIII. Members of this protein class play key roles during normal processes that occur during development, tissue morphogenesis, and wound healing. As key components of basement membranes in organs and tissues, they also participate in selective filtration of biological fluids, in establishing cellular barriers, and in modulation of angiogenesis. The ability to perform these functions is provided both by the features of the protein cores as well as by the unique properties of heparan sulfate, which is assembled as a polymer of N-acetylglucosamine and glucuronic acid and modified by specific enzymes to generate specialized biologically active structures. This article discusses the structures and functions of this amazing family of proteoglycans and provides a platform for further study of the individual members.
Collapse
Affiliation(s)
| | - Mary C. Farach-Carson
- Department of Biological Sciences, University of Delaware, Newark, DE 19707 USA
- Present Address: Department of Biochemistry and Cell Biology, Weiss School of Natural Sciences, Rice University, MS-102, P.O. Box 1892, Houston, TX 77251-1892 USA
| | - Daniel D. Carson
- Present Address: Department of Biochemistry and Cell Biology, Weiss School of Natural Sciences, Rice University, MS-102, P.O. Box 1892, Houston, TX 77251-1892 USA
| |
Collapse
|
43
|
Lambaerts K, Wilcox-Adelman SA, Zimmermann P. The signaling mechanisms of syndecan heparan sulfate proteoglycans. Curr Opin Cell Biol 2009. [DOI: 10.1016/j.ceb.2009.05.002 doi:dx.doi.org] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
|
44
|
Kirn-Safran C, Farach-Carson MC, Carson DD. Multifunctionality of extracellular and cell surface heparan sulfate proteoglycans. Cell Mol Life Sci 2009. [DOI: 10.1007/s00018-009-0096-1 doi:dx.doi.org] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|
45
|
The signaling mechanisms of syndecan heparan sulfate proteoglycans. Curr Opin Cell Biol 2009; 21:662-9. [PMID: 19535238 DOI: 10.1016/j.ceb.2009.05.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 05/13/2009] [Indexed: 01/01/2023]
Abstract
Syndecans are membrane proteins controlling cell proliferation, differentiation, adhesion, and migration. Their extracellular domains bear versatile heparan sulfate chains that provide structural determinants for syndecans to function as coreceptors or activators for molecules like growth factors and constituents of the matrix. Syndecans also signal via their protein cores and their conserved transmembrane and cytoplasmic domains. The direct interactions and signaling cascades they support are becoming better characterized. These interactions are regulated by phosphorylation, induced clustering and shedding of the syndecan extracellular domain. Moreover evidence is emerging that syndecans concentrate in unconventional lipid domains and might govern novel vesicular trafficking pathways. Here we focus on recent findings that refine our understanding of the complex structure-function relationships of these cellular effectors.
Collapse
|
46
|
Jurjus RA, Liu Y, Pal-Ghosh S, Tadvalkar G, Stepp MA. Primary dermal fibroblasts derived from sdc-1 deficient mice migrate faster and have altered alphav integrin function. Wound Repair Regen 2009; 16:649-60. [PMID: 19128260 DOI: 10.1111/j.1524-475x.2008.00423.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT The goal of this study is to determine whether dermal fibroblasts lacking syndecan-1 (sdc1) show differences in integrin expression and function that could contribute to the delayed skin and corneal wound healing phenotypes seen in sdc-1 null mice. Using primary dermal fibroblasts, we show that after 3 days in culture no differences in alpha-smooth muscle actin were detected but sdc-1 null cells expressed significantly more alphav and beta1 integrin than wildtype (wt) cells. Transforming growth factor beta1 (TGFbeta1) treatment at day 3 increased alphav- and beta1-integrin expression in sdc-1 null cells at day 5 whereas wt cells showed increased expression only of alphav-integrin. Using time-lapse studies, we showed that the sdc-1 null fibroblasts migrate faster than wt fibroblasts, treatment with TGFbeta1 increased these migration differences, and treatment with a TGFbeta1 antagonist caused sdc-1 null fibroblasts to slow down and migrate at the same rate as untreated wt cells. Cell spreading studies on replated fibroblasts showed altered cell spreading and focal adhesion formation on vitronectin and fibronectin-coated surfaces. Additional time lapse studies with beta1- and alphav-integrin antibody antagonists, showed that wt fibroblasts expressing sdc-1 had activated integrins on their surface that impeded their migration whereas the null cells expressed alphav-containing integrins which were less adhesive and enhanced cell migration. Surface expression studies showed increased surface expression of alpha2beta1 and alpha3beta1 on the sdc-1 null fibroblasts compared with wt fibroblasts but no significant differences in surface expression of alpha5beta1, alphavbeta3, or alphavbeta5. Taken together, our data indicates that sdc-1 functions in the activation of alphav-containing integrins and support the hypothesis that impaired wound healing phenotypes seen in sdc-1 null mice could be due to integrin-mediated defects in fibroblast migration after injury.
Collapse
Affiliation(s)
- Rosalyn A Jurjus
- Department of Anatomy and Regenerative Biology, George Washington University Medical School, 2300 I Street NW, Washington, DC 20037, USA
| | | | | | | | | |
Collapse
|
47
|
Kharabi Masouleh B, Ten Dam GB, Wild MK, Seelige R, van der Vlag J, Rops AL, Echtermeyer FG, Vestweber D, van Kuppevelt TH, Kiesel L, Götte M. Role of the heparan sulfate proteoglycan syndecan-1 (CD138) in delayed-type hypersensitivity. THE JOURNAL OF IMMUNOLOGY 2009; 182:4985-93. [PMID: 19342678 DOI: 10.4049/jimmunol.0800574] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The cell surface heparan sulfate proteoglycan syndecan-1 (CD138) modulates the activity of chemokines, cytokines, integrins, and other adhesion molecules which play important roles in the regulation of inflammation. We have previously shown that syndecan-1-deficient murine leukocytes display increased interactions with endothelial cells and increased diapedesis in vivo and in vitro. In this study, we demonstrate that syndecan-1 has an important function as a negative modulator in the murine contact allergy model of oxazolone-mediated delayed-type hypersensitivity (DTH). Following elicitation of the DTH response, syndecan-1-deficient mice showed an increase in leukocyte recruitment, resulting in an increased and prolonged edema formation. Expression of the cytokines TNF-alpha and IL-6 of the chemokines CCL5/RANTES and CCL-3/MIP-1alpha and of the adhesion molecule ICAM-1 were significantly increased in syndecan-1-deficient compared with wild-type mice. In wild-type mice, syndecan-1 mRNA and protein expression was reduced during the DTH response. The differentially increased adhesion of syndecan-1-deficient leukocytes to ICAM-1 was efficiently inhibited in vitro by CD18-blocking Abs, which emerges as one mechanistic explanation for the anti-inflammatory effects of syndecan-1. Collectively, our results show an important role of syndecan-1 in the contact DTH reaction, identifying syndecan-1 as a novel target in anti-inflammatory therapy.
Collapse
Affiliation(s)
- Behzad Kharabi Masouleh
- Department of Gynecology and Obstetrics, Medical Center, University of Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nikolova V, Koo CY, Ibrahim SA, Wang Z, Spillmann D, Dreier R, Kelsch R, Fischgräbe J, Smollich M, Rossi LH, Sibrowski W, Wülfing P, Kiesel L, Yip GW, Götte M. Differential roles for membrane-bound and soluble syndecan-1 (CD138) in breast cancer progression. Carcinogenesis 2009; 30:397-407. [PMID: 19126645 DOI: 10.1093/carcin/bgp001] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The heparan sulfate proteoglycan syndecan-1 (Sdc1) modulates cell proliferation, adhesion, migration and angiogenesis. Proteinase-mediated shedding converts Sdc1 from a membrane-bound coreceptor into a soluble effector capable of binding the same ligands. In breast carcinomas, Sdc1 overexpression correlates with poor prognosis and an aggressive phenotype. To distinguish between the roles of membrane-bound and shed forms of Sdc1 in breast cancer progression, human MCF-7 breast cancer cells were stably transfected with plasmids overexpressing wild-type (WT), constitutively shed and uncleavable forms of Sdc1. Overexpression of WT Sdc1 increased cell proliferation, whereas overexpression of constitutively shed Sdc1 decreased proliferation. Fibroblast growth factor-2-mediated mitogen-activated protein kinase signaling was reduced following small-interfering RNA (siRNA)-mediated knockdown of Sdc1 expression. Constitutively, membrane-bound Sdc1 inhibited invasiveness, whereas soluble Sdc1 promoted invasion of MCF-7 cells into matrigel matrices. The latter effect was reversed by the matrix metalloproteinase inhibitors N-isobutyl-N-(4-methoxyphenylsufonyl) glycyl hydroxamic acid and tissue inhibitor of metalloproteinase (TIMP)-1. Affymetrix microarray analysis identified TIMP-1, Furin and urokinase-type plasminogen activator receptor as genes differentially regulated in soluble Sdc1-overexpressing cells. Endogenous TIMP-1 expression was reduced in cells overexpressing soluble Sdc1 and increased in those overexpressing the constitutively membrane-bound Sdc1. Moreover, E-cadherin protein expression was downregulated in cells overexpressing soluble Sdc1. Our results suggest that the soluble and membrane-bound forms of Sdc1 play different roles at different stages of breast cancer progression. Proteolytic conversion of Sdc1 from a membrane-bound into a soluble molecule marks a switch from a proliferative to an invasive phenotype, with implications for breast cancer diagnostics and potential glycosaminoglycan-based therapies.
Collapse
Affiliation(s)
- Viktoriya Nikolova
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Association of loss of epithelial syndecan-1 with stage and local metastasis of colorectal adenocarcinomas: an immunohistochemical study of clinically annotated tumors. BMC Cancer 2008; 8:185. [PMID: 18590537 PMCID: PMC2459187 DOI: 10.1186/1471-2407-8-185] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Accepted: 06/30/2008] [Indexed: 11/12/2022] Open
Abstract
Background Syndecan-1 is a transmembrane proteoglycan with important roles in cell-cell and cell-extracellular matrix adhesion and as a growth factor co-receptor. Syndecan-1 is highly expressed by normal epithelial cells and loss of expression has been associated with epithelial-mesenchymal transition and the transformed phenotype. Loss of epithelial syndecan-1 has been reported in human colorectal adenocarcinomas, but whether this has prognostic significance remains undecided. Here we have examined syndecan-1 expression and its potential prognostic value with reference to a clinically annotated tissue microarray for human colon adenocarcinomas. Methods Syndecan-1 expression was examined by immunohistochemistry of a tissue microarray containing cores from 158 colorectal adenocarcinomas and 15 adenomas linked to a Cleveland Clinic, IRB-approved database with a mean clinical follow-up of 38 months. The Kaplan-Meier method was used to analyze the relationship between syndecan-1 expression and patient survival. Potential correlations between syndecan-1 expression and the candidate prognostic biomarker fascin were examined. Results Syndecan-1 is expressed at the basolateral borders of normal colonic epithelial cells. On adenocarcinoma cells, syndecan-1 was present around cell membranes and in cytoplasm. In 87% of adenocarcinomas, syndecan-1 was decreased or absent; only 13% of patients had stained for syndecan-1 on more than 75% of tumor cells. Decreased syndecan-1 correlated with a higher TNM stage and lymph node metastasis and was more common in males (p = 0.042), but was not associated with age, tumor location or Ki67 index. Reduced tumor syndecan-1 staining also correlated with upregulation of stromal fascin (p = 0.016). Stromal syndecan-1 was observed in 16.6% of tumors. There was no difference in survival between patients with low or high levels of either tumor or stromal syndecan-1. Conclusion Syndecan-1 immunoreactivity was decreased in the majority of human colon adenocarcinomas in correlation with TNM stage and metastasis to local lymph nodes. In a small fraction of adenocarcinomas, syndecan-1 was upregulated in the local stroma. Syndecan-1 expression status did not correlate with patient survival outcomes. Combined analysis of syndecan-1 in relation to a potential prognostic biomarker, fascin, identified that loss of tumor syndecan-1 correlated significantly with strong stromal fascin staining.
Collapse
|
50
|
Stepp MA, Liu Y, Pal-Ghosh S, Jurjus RA, Tadvalkar G, Sekaran A, Losicco K, Jiang L, Larsen M, Li L, Yuspa SH. Reduced migration, altered matrix and enhanced TGFbeta1 signaling are signatures of mouse keratinocytes lacking Sdc1. J Cell Sci 2007; 120:2851-63. [PMID: 17666434 DOI: 10.1242/jcs.03480] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have reported previously that syndecan-1 (Sdc1)-null mice show delayed re-epithelialization after skin and corneal wounding. Here, we show that primary keratinocytes obtained from Sdc1-null mice and grown for 3-5 days in culture are more proliferative, more adherent and migrate more slowly than wt keratinocytes. However, the migration rates of Sdc1-null keratinocytes can be restored to wild-type levels by replating Sdc1-null keratinocytes onto tissue culture plates coated with fibronectin and collagen I, laminin (LN)-332 or onto the matrices produced by wild-type cells. Migration rates can also be restored by treating Sdc1-null keratinocytes with antibodies that block alpha6 or alphav integrin function, or with TGFbeta1. Antagonizing either beta1 integrin function using a function-blocking antibody or TGFbeta1 using a neutralizing antibody reduced wild-type keratinocyte migration more than Sdc1-null keratinocyte migration. Cultures of Sdc1-null keratinocytes accumulated less collagen than wild-type cultures but their matrices contained the same amount of LN-332. The Sdc1-null keratinocytes expressed similar total amounts of eight different integrin subunits but showed increased surface expression of alphavbeta6, alphavbeta8, and alpha6beta4 integrins compared with wild-type keratinocytes. Whereas wild-type keratinocytes increased their surface expression of alpha2beta1, alphavbeta6, alphavbeta8, and alpha6beta4 after treatment with TGFbeta1, Sdc1-null keratinocytes did not. Additional data from a dual-reporter assay and quantification of phosphorylated Smad2 show that TGFbeta1 signaling is constitutively elevated in Sdc1-null keratinocytes. Thus, our results identify TGFbeta1 signaling and Sdc1 expression as important factors regulating integrin surface expression, activity and migration in keratinocyte and provide new insight into the functions regulated by Sdc1.
Collapse
Affiliation(s)
- Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University Medical School, Washington, DC 20037, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|